MIC-1 Receptor and Uses Thereof

The present invention relates to the newly identified MIC-1 binding receptor, GFRAL. In vitro bioassays, for testing affinity and potency of GFRAL ligand, such as MIC- or MIC-1 variants, are provided.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
FIELD OF THE INVENTION

The present invention relates to assays for screening or evaluating GFRAL ligands, such as MIC-1 compounds. The present invention also relates to cell lines for determining the activity of GFRAL ligands.

BACKGROUND OF THE INVENTION

Weight and appetite control are complex. Its dysregulation can lead to obesity or anorexia.

Obesity is believed to be one of the principal factors responsible for the development of insulin resistance, impaired glucose homeostasis. There has been proposed several mechanisms linking obesity with type 2 diabetes such as ectopic lipid accumulation, low-grade chronic inflammation, endothelial dysfunction, leptin resistance, gut flora, impaired energy expenditure just to mention some. Interestingly, while the relative contributions from each of these factors remain uncertain, it is believed that many of these factors are either induced or accelerated with obesity. Thus, the most direct approach to prevent and treat newly diagnosed T2 diabetics is to reduce food intake and obesity.

At the other extreme, anorexia/cachexia is most commonly due to late stage cancer, where it is believed that tumor or stromal cell derived molecules disturb the control of appetite and weight, leading to wasting, debility and often death.

MIC-1 was first reported as a new member of the transforming TGF-β super-family in 1997, and it was named Macrophage Inhibitory Cytokine-1 (MIC-1). See Bootcov, M. R. et al. MIC-1, a novel macrophage inhibitory cytokine, is a divergent member of the TGF-β superfamily cluster. Proc. Natl. Acad. Sci. USA 94, 11514-11519 (1997).

Data in literature show that MIC-1 is not expressed under basal conditions but may be induced by inflammation, injury or malignancy. MIC-1 is overexpressed in many cancers, including those of the prostate, colon, pancreas and breast. In individuals with advanced cancer, serum MIC-1 can rise from a mean of 0.45 ng/ml to 5-50 ng/ml or higher. Tumor overproduction of MIC-1 and the correlation of serum MIC-1 levels with weight loss (in both animal models and in individuals with prostate cancer) suggest that MIC-1 is involved in the pathogenesis of cancer anorexia and weight loss and is perhaps involved in other cachectic conditions, such as those that are associated with renal and cardiac failure. The weight loss in animal model was reversed by treatment with MIC-1 antibody. MIC-1 also induced weight loss in massively obese leptin-deficient ob/ob mice. The studies in the literature show that MIC-1 is a regulator of appetite and weight, and it could be a therapeutic target for cancer anorexia, weight loss, obesity, and other conditions that are responsive to modulating the activity of MIC-1. See, for example, Breit S N et al. Tumor-induced anorexia and weight loss are mediated by the TGF-β superfamily cytokine MIC-1. Nat Med 2007; 13: 1333-1340. However, MIC-1's mechanism of action is not known.

SUMMARY OF THE INVENTION

In one aspect, the invention provides a method or an assay for testing or evaluating the activity of a GFRAL ligand. In one aspect, the invention provides a method or an assay for testing or evaluating the activity of a MIC-1 compound.

In a further aspect, the invention provides a cell-based assay or a cell line for detecting or evaluating the activity of a GFRAL ligand, such as a MIC-1 compound, wherein the cell expresses GFRAL ligand receptor on cell surface. The cell-based assay could be a proliferation assay, cytotoxicity assay, gene expression assay, a signal transduction assay, etc.

In another aspect, the invention provides a method or an assay for detecting or evaluating the binding affinity and selectivity of a GFRAL ligand, such as a MIC-1 compound, to GFRAL.

Also or alternatively, the invention provides a kit for measuring the activity and selectivity of a GFRAL ligand, such as a MIC-1 compound.

Also or alternatively, the invention provides a kit for measuring the binding affinity and selectivity of a GFRAL ligand, such as a MIC-1 compound.

DESCRIPTION OF THE DRAWINGS

FIG. 1 shows that administration of mAb26 inhibited wtMIC-1's in vivo activity on suppressing food intake in sprague dawley male rats. Accumulated food intake was measured over 24 hrs in rats after SC administration of 200 μg/kg of wtMIC-1. Open Square: vehicle group pre-treated with isotype antibody. Black circle: wtMIC-1 group pre-treated with isotype antibody (control); Black triangle: wtMIC-1 group pre-treated with mAb26. Vehicle and wtMIC-1 was administrated SC and antibodies was administrated IP.

FIG. 2 shows that mAb26 blocked binding of wtMIC-1 to immobilized hGFRAL(ECR)-Fab in SPR in a dose-dependent manner.

FIG. 3 shows that mAb26 blocked binding of wtMIC-1 to immobilized mGFRAL(ECR)-Fab in SPR in a dose-dependent manner.

FIG. 4 shows that RET receptor tyrosine kinase is the co-binder of MIC-1 and GFRAL. In the presence of wtMIC-1, Ret-Fc specifically bound to hGFRAL(ECR) but not to GFRA1(ECR). Ret-Fc did not bind to wtMIC-1, nor to hGFRAL(ECR) in the absence of wtMIC-1.

FIG. 5 shows that wtMIC-1 specifically bound to GFRAL, but not to the four receptors of GDNF family (GDNF, NRTN, ARTN and PSPN) in SPR.

FIG. 6 shows that GFRAL domain(s) for MIC-1 binding interaction is located within C1C2 domains of the extracellular region of hGFRAL. wtMIC-1 strongly bound to hGFRAL_C1C2 at 1 nM in SPR; wtMIC-1 weakly bound to hGFRAL_C2 or hGFRAL_C2C3 at 1000 nM in SPR.

FIG. 7 shows that C3 domain of the extracellular region of GFRAL is necessary for Ret to bind to the MIC-1/GFRAL complex for forming MIC-1/GFRAL/Ret ternary complex, although GFRAL C3 is dispensable for MIC-1 binding to GFRAL.

FIG. 8 shows the relative binding of Ret-Fc in the FACS dot-plot drawings of FIG. 7.

FIG. 9 shows that wtMIC-1 induced ERK1/2 phosphorylation in PC12 cells co-transfected with and expressed full length hGFRAL and full length hRET51.

FIG. 10 shows that wtMIC-1 induced ERK1/2 phosphorylation in HEK293 cells co-transfected with and expressed full length hGFRAL and full length hRET51.

FIG. 11 shows that MIC-1 only induced ERK phosphorylation in cells co-transfected with GFRAL and RET, but not in cells transfected with GFRAL only, or in cells transfected with RET only.

FIG. 12 shows that MIC-1 only induced ERK phosphorylation in cells co-transfected with hRET51 and hGFRAL, not in cells co-transfected with hRET43 and hGFRAL or hRET9 and hGFRAL

FIG. 13 showed that MIC-1 can also induced ERK phosphorylation in cells co-transfected with rat GFRAL and rat RET, or cells co-transfected with cynomolgus (cyno) GFRAL and cyno RET.

FIG. 14 showed that Hedgehog, MEF2 and MAPK/ERK signaling pathways were regulated upon MIC-1 treatment, as evidenced by up-regulation of GLI, MEF2 and SRE derived luciferase reporter activities respectively when tested by Cignal 45-pathway Reporter Array.

FIG. 15 shows that wtMIC-1 bound to BHK21 cells co-transfected and expressed full length hGFRAL and full length hRET in a dose-dependent manner.

FIG. 16 shows that wtMIC-1 induced both phosphorylation of ERK and AKT in BHK21-hGFRAL-IRES-hRET stable cells. Western blot showed the protein level change of phosphorylation of ERK1/2 and AKT1/2/3 after wtMIC-1 induction.

FIG. 17 shows that wtMIC-1 induced ERK1/2 phosphorylation in BHK21-hGFRAL-IRES-hRET stable cells in a dose-dependent manner.

FIG. 18 shows that wtMIC-1 induced AKT1/2/3 phosphorylation in BHK21-hGFRAL-IRES-h RET stable cells in a dose-dependent manner.

FIG. 19 shows that wtMIC-1 induced ERK1/2 phosphorylation in BHK21-hGFRAL-IRES-hRET-SRE-Luc stable cells cells in a dose-dependent manner.

FIG. 20 shows that wtMIC-1 activated luciferase in BHK21-hGFRAL-IRES-hRET-SRE-Luc stable cells in a dose-dependent manner

BRIEF DESCRIPTION OF THE SEQUENCE LISTING

SEQ ID NO: 1 gives the DNA sequence of full length human GFRAL (hGFRAL). Residues 88-1272 is the sequence of coding sequence (CDS).

SEQ ID NO: 2 gives the amino acid sequence of the full length hGFRAL(hGFRAL(FL)). Residues 1-18 is the sequence of the signal peptide. Residues 19-351 is the sequence of the extracellular region (ECR). Residues 352-371 is the sequence of the transmembrane domain. Residues 372-394 is the sequence of the intracellular region (ICR).

SEQ ID NO: 3 gives the DNA sequence of mouse GFRAL isoform 1 (mGFRAL). Residues 196-1377 is the sequence of CDS.

SEQ ID NO: 4 gives the amino acid sequence of mGFRAL isoform1. Residues 1-19 is the sequence of the signal peptide. Residues 20-349 is the sequence of the extracellular region (ECR). Residues 350-370 is the sequence of the transmembrane domain. Residues 371-393 is the sequence of the intracellular region (ICR).

SEQ ID NO: 5 gives the amino acid sequence of human RET isoform 51 (hRET51). Residues 1-28 is the sequence of the signal peptide. Residues 29-636 is the sequence of the extracellular region (ECR). Residues 637-667 is the sequence of the transmembrane domain. Residues 668-1114 is the sequence of the intracellular region (ICR).

SEQ ID NO: 6 gives the amino acid sequence of hGFRAL_ECR-LC. Residues 1-16 is the sequence of the CD33 signal peptide. Residues 17-348 is the sequence of ECR of hGFRAL. Residues 349-358 is the sequence of the GGGGSGGGGS linker. Residues 359-577 is the sequence of mouse anti-TNP light chain (LC).

SEQ ID NO: 7 gives the amino acid sequence of hGFRAL_ECR-hIgG1.1_Fc. Residues 1-16 is the sequence of the CD33 signal peptide. Residues 17-340 is the sequence of ECR of hGFRAL. Residues 341-575 is the sequence of human IgG1.1_Fc (hIgG1.1_Fc).

SEQ ID NO: 8 gives the amino acid sequence of mGFRAL_ECR-LC. Residues 1-16 is the sequence of the CD33 signal peptide. Residues 17-345 is the sequence of ECR of mGFRAL. Residues 346-356 is the sequence of the GGGGSGGGGS linker. Residues 357-575 is the sequence of mouse anti-TNP light chain (LC).

SEQ ID NO: 9 gives the amino acid sequence of hGFRAL_C1-LC. Residues 1-16 is the sequence of the CD33 signal peptide. Residues 17-127 is the sequence of the C1 domain in ECR of hGFRAL. Residues 128-137 is the sequence of the GGGGSGGGGS linker. Residues 138-356 is the sequence of mouse anti-TNP light chain (LC).

SEQ ID NO: 10 gives the amino acid sequence of hGFRAL_C2-LC. Residues 1-16 is the sequence of the CD33 signal peptide. Residues 17-105 is the sequence of the C2 domain in ECR of hGFRAL. Residues 106-115 is the sequence of the GGGGSGGGGS linker. Residues 116-334 is the sequence of mouse anti-TNP light chain (LC).

SEQ ID NO: 11 gives the amino acid sequence of hGFRAL_C3-LC. Residues 1-16 is the sequence of the CD33 signal peptide. Residues 17-148 is the sequence of the C2 domain in ECR of hGFRAL. Residues 149-158 is the sequence of the GGGGSGGGGS linker. Residues 159-377 is the sequence of mouse anti-TNP light chain (LC).

SEQ ID NO: 12 gives the amino acid sequence of hGFRAL_C1C2-LC. Residues 1-16 is the sequence of the CD33 signal peptide. Residues 17-216 is the sequence of the C1 and C2 domains in ECR of hGFRAL. Residues 217-226 is the sequence of the GGGGSGGGGS linker. Residues 227-445 is the sequence of mouse anti-TNP light chain (LC).

SEQ ID NO: 13 gives the amino acid sequence of hGFRAL_C2C3-LC. Residues 1-16 is the sequence of the CD33 signal peptide. Residues 17-237 is the sequence of the C2 and C3 domains in ECR of hGFRAL. Residues 238-247 is the sequence of the GGGGSGGGGS linker. Residues 248-466 is the sequence of mouse anti-TNP light chain (LC).

SEQ ID NO: 14 gives the amino acid sequence of HC_TM (chimeric protein consisted of mouse anti-TNF fab heavy chain and PDGFR transmembrane region). Residues 1-18 is the sequence of the signal peptide of mouse Ig heavy chain. Residues 19-236 is the sequence of mouse anti-TNP fab heavy chain (HC). Residues 237-285 is the sequence of a fragment of human PDGFRB containing the transmembrane region. Residues 257-277 is the sequence of the PDGFRB transmembrane region (TM).

SEQ ID NO: 15 gives the amino acid sequence of HC_HPC4 (chimeric protein consisted of mouse anti-TNF fab heavy chain and HPC4 tag). Residues 1-18 is the sequence of the signal peptide of mouse Ig heavy chain. Residues 19-236 is the sequence of mouse anti-TNP fab heavy chain (HC). Residues 237-241 is the sequence of GGGGS linker. Residues 242-253 is the sequence of HPC4 tag (HPC4).

SEQ ID NO: 16 gives the amino acid sequence of wild type human MIC-1 (wtMIC-1).

SEQ ID NO: 17 gives the amino acid sequence of the full length human RET43.

SEQ ID NO: 18 gives the amino acid sequence of the full length human RET9.

SEQ ID NO: 19 gives the amino acid sequence of the full length mouse RET.

SEQ ID NO: 20 gives the amino acid sequence of the full length cyno GFRAL.

SEQ ID NO: 21 gives the amino acid sequence of the full length cyno RET.

SEQ ID NO: 22 gives the amino acid sequence of the full rat GFRAL.

SEQ ID NO: 23 gives the amino acid sequence of the full length rat RET.

DESCRIPTION OF THE INVENTION

The inventors of the present application surprisingly found that GFRAL is the cell surface receptor that mediates the in vivo activities of MIC-1. In particular, GFRAL binds to MIC-1 and then the complex of GFRAL and MIC-1 binds to RET to mediate activation of MIC-1.

In one aspect, the invention provides a cell-based potency assay to test the biological activity of a GFRAL ligand, comprising contacting an indicator cell with a test sample that comprises the GFRAL ligand, and then detecting a biological response of the indicator cell. The indicator cell expresses on the cell surface a GFRAL ligand binding receptor comprising a GFRAL ligand binding segment derived from GFRAL. In one embodiment, the GFRAL ligand is a MIC-1 compound, and GFRAL ligand binding receptor is MIC-1 binding receptor that comprises a MIC-1 binding segment derived from GFRAL.

As used herein, “potency” is a measure of substance's biological activity of a substance that elicits or produces a defined biological response. In the pharmaceutical area, potency is a measure of the biological activity of a drug product that produces a defined clinical effect.

As used herein, “biological activity” is the ability of a molecule to effect a change in a biological process. Here, biological activity, bioactivity, functional activity, and function are interchangeable. Characterization of a biological product includes the determination of physicochemical properties, biological activity, immunochemical properties, purity and impurities, etc. Cell culture based or cell-based potency assays are often the preferred format for determining the biological activity of a biological product, since they can measure the biological response elicited by the product and can generate results within a relatively short period of time, comparing with animal based assays. Also, lots of cell-based potency assays have defined correlation with the drug product's mechanism of action. Thus, such assays are widely used and required by drug administration authorities for drug registration and release.

Cells are living entities, representing biological systems that possess many of the important in vivo characteristics that make them useful for measuring biological activity. Such cell-based assays is capable of being used for characterization, lot release, in-process and stability testing for drugs. Such assays can provide a basis for assessing product comparability before and after manufacturing changes. They can evaluate product stability (e.g., expiry dating), and control clinical dosing consistency. Other uses of cell-based assays include qualification of internal reference materials; characterization of process intermediates, formulations, and degradation products, etc.

In one aspect of the invention, indicator cell line is from a cell lineage close to the cell or tissue type targeted by MIC-1 compounds in vivo. In another aspect of the invention, it is a cell line expressing an appropriate MIC-1 compound binding receptor on the cell surface, either endogenously or exogenously via transfection. In one embodiment, the indicator cell is derived from neuron cells. In a further embodiment, the indicator cell is derived from hypothalamus or brain stem. In yet another embodiment, the indicator cell is derived from arcuate nucleus (ARC), paraventricular nucleus (PVN), area postrema (AP), or nucleus tractus solitarus (NTS). In a further embodiment, the indicator cells are derived from neuron cells, e.g. cells derived from ARC, PVN, AP, NTS, express the MIC-1 binding receptor endogenously. In yet another embodiment, indicator cell is derived from arcuate nucleus (ARC), paraventricular nucleus (PVN), area postrema (AP), or nucleus tractus solitarus (NTS) express the MIC-1 binding receptor exogenously via transfection or transformation with a construct or vector. In another embodiment, the indicator cell is a cell transfected or transformed with a construct or vector to exogenously express the MIC-1 binding receptor on the cell surface. The transformed or transfected cells can be prokaryotes, or eukaryotes. Prokaryotes include gram negative bacteria and gram positive bacteria. The indicator cell can also be yeast cells or animal cells. In an embodiment, the animal cell is originated from non-mammalian animal, e.g., insects or birds. In another embodiment, the animal cell is originated from mammalian animal, e.g., human, primates, and rodents. In a further embodiment, the indicator cell is BHK21, PC-12, HEK293, 3T3-L1, CHO, HTC-116, PC3, or Caco2.

“Construct” is interchangeable with “vector” in this document, which refers to an artificially assembled DNA segment to be transferred into a cell line, target tissue, or animal. Construct or vectors, as used herein, include plasmid, virus, bacteriophage, integratable DNA fragments (i.e., fragments integratable into the host genome by genetic recombination), and other vehicles which enable the integration of DNA fragments comprising a gene or a nucleic acid sequence of interest. Typically, the construct comprises control elements and a gene or a nucleic acid sequence encoding MIC-1 binding receptor. Generally, the control elements include a promoter system, an operator to control the level of mRNA expression, a sequence encoding a ribosome binding site, a sequence terminating transcription and translation, etc.

The term “biological response” as used herein means a response of the indicator cell elicited by the molecules to be tested, e.g., GFRAL ligand (such as MIC-1 compound). Biological responses include any responses related to for example cell proliferation, validity, growth arrest, cell death (e.g. apoptosis), motility, morphology, toxicity, gene transcription (e.g. reporter genes), protein expression, cytokine release, phosphorylation of a kinase, or activation or inactivation of any components of a signaling pathway, metabolism, etc.

In one embodiment, the biological response is up-regulating or down-regulating a gene transcription. In a further embodiment, the up-regulated gene is c-fos. In another embodiment, the up-regulated gene is a gene of an appetite regulating neuro-peptide. In a further embodiment, the appetite regulating neuro-peptide is POMC (pro-opiomelanocortin), CART (cocaine and amphetamine regulated transcript), NPY (neuropeptide Y), or AGRP (agouti-related protein). In yet a further embodiment, the mRNA of POMC and/or CART is increased. In yet another embodiment, the mRNA of NYP and/or AGRP is decreased. In yet another embodiment, the up-regulated gene transcription is an indicator of signaling pathway activation. In a further embodiment, transcription of GLI, MEF2, SRE, FOXO is upregulated.

In one embodiment, the biological response is related to signal transduction. Cell signaling or signal transduction is a part of a communication system that governs cellular activities and coordinates cell actions. Through such communication systems including signal transduction, cells are able to perceive and respond to the changes in the surrounding microenvironment.

In one embodiment, the signal transduction related biological response is an increase or decrease of phosphorylation of a protein kinase. In a further embodiment, the signal transduction related biological response is an increase or decrease of phosphorylation of RET, ERK1/2, AKT1/2/3, STAT(s), etc. One major mechanism for signal transduction in animals involves protein phosphorylation. Protein phosphorylation involves the action of protein kinase, an enzyme that transfers a phosphate group from a phosphate donor onto an acceptor amino acid in a substrate protein. Protein kinases can be classified based on the acceptor amino acid specificity. The two most well characterized types of protein kinases are protein serine/threonine kinases (a protein kinase with a protein alcohol group as acceptor) and protein tyrosine kinases (a protein kinases with a protein phenolic group as acceptor).

Various assays have been developed for measuring the phosphorylation of serine/threonine kinases and tyrosine kinases. Some of these assays measure the ability of a tyrosine kinase or serine/threonine kinase to phosphorylate a synthetic substrate polypeptide. See Pike, L., Methods of Enzymology 146:353-362 (1987) and Hunter, Journal of Biological Chemistry 257(9):4843-4848 (1982). Wang et al., Journal of Biological Chemistry 267(24):17390-17396 [1992]. Such assays can use radioactive labels. The discovery of phospho-specific antibodies against phosphorylated of tyrosine/serine/threonine residues or against a specific phosphorylated protein kinase enables immunology assays measuring phosphorylation of protein kinases, such as ELISA.

In another embodiment, the biological response detected is an increase or decrease of the level of a second messenger.

Except for protein phosphorylation, second messengers usually are also involved in intracellular signal transduction. Second messengers are signaling molecules released by the cell to trigger physiological changes such as proliferation, differentiation, migration, survival, and apoptosis, etc. Examples of second messenger molecules include cyclic AMP, cyclic GMP, inositol triphosphate, diacylglycerol, and calcium. In one embodiment, the second messenger is coupled to phosphorylation of a protein kinase. For example, Ras and GTP link with the Mitogen Activated Protein Kinase (MAPK) cascade.

GFRAL is the abbreviation of “GDNF family receptor alpha like”. Human GFRAL was described in WO 03/076569. Mouse GFRAL was described by Li Z, et al. in Identification, expression and functional characterization of the GRAL gene, J Neurochem, 2005 October. Sequence analysis suggests that GFRAL is a remote homolog of GFRα1-4 (GDNF family receptor alpha 1-4).

“GFRAL ligand” as used herein means a ligand (a compound) that binds to the GFRAL receptor. “GFRAL ligand” can be an antagonist or against of GFRAL receptor. In one embodiment, GFRAL ligand is an antibody, e.g. an antagonistic antibody or agonistic antibody. In another embodiment, GFRAL ligand is a MIC-1 compound.

The term “MIC-1 compound” as used herein means MIC-1 of mammalian origin, such as human, monkey, rat, mouse, etc., and recombinant MIC-1, such as recombinant human, monkey, rat, mouse MIC-1, and a MIC-1 variant. A MIC-1 variant means a variant that retains MIC-1 activity. MIC-1 variant could be a truncated MIC-1, a MIC-1 analogue or a MIC-1 derivative. As used herein “MIC-1” and “MIC-1 compound” are interchangeable. A truncated MIC-1 is a fragment of wild type MIC-1. A MIC-1 analogue means a modified MIC-1 wherein one or more amino acid residues of wild type MIC-1 have been substituted with other natural or unnatural amino acid residues and/or wherein one or more natural or unnatural amino acid residues have been deleted from wild type MIC-1 and/or wherein one or more natural or unnatural amino acid residues have been added to wild type MIC-1 and any combinations thereof. MIC-1 derivative means a chemically modified wild type MIC-1 with or without substituting, adding or deleting one or more natural or unnatural amino acid residues, wherein at least one substituent is not present in wild type MIC-1. Typical modifications include amides, carbohydrates, alkyl groups, acyl groups, esters, PEGylations and the like.

In some embodiments, the MIC-1 compound has at least 80% sequence identity or homology with wild type MIC-1. In some embodiments, the MIC-1 compound has at least 10% of the wild type MIC-1 activity. “Identity” and “homology” are interchangeable in this document. In some embodiments, the MIC-1 has at least 80% identity with wild type human MIC-1 and at least 10% of the activity of the wild type human MIC-1. In a further embodiment, MIC-1 represents a MIC-1 compound comprising an amino acid sequence having at least 90% identity to the amino acid sequence of human MIC-1. In further embodiments, MIC-1 has at least 80%, at least 85%, at least 95%, at least 96%, at least 97%, at least 98% or at least 99% identity with human MIC-1. In further embodiments, MIC-1 has at least 10%, at least 20%, at least 30%, at least 40%, at least 50%, at least 60%, at least 70%, at least 80%, or at least 90% of the MIC-1 activity of human MIC-1.

“GFRAL variant” as used herein means a GFRAL variant that retains MIC-1 binding affinity of wtGFRAL. A GFRAL variant could be a truncated GFRAL, a GFRAL analogue or a GFRAL derivative. A truncated GFRAL is a fragment of wild type GFRAL. In one embodiment, a truncated GFRAL comprises C1 and C2 domains of GFRAL. A GFRAL analogue means a modified GFRAL wherein one or more amino acid residues of wild type GFRAL have been substituted with other natural or unnatural amino acid residues and/or wherein one or more natural or unnatural amino acid residues have been deleted from wild type GFRAL and/or wherein one or more natural or unnatural amino acid residues have been added to wild type GFRAL and any combinations thereof. GFRAL derivative means a chemically modified wild type GFRAL with or without substituting, adding or deleting one or more natural or unnatural amino acid residues, wherein at least one substituent is not present in wild type GFRAL. Typical modifications include amides, carbohydrates, alkyl groups, acyl groups, esters, PEGylations and the like.

In some embodiments, the GFRAL variant has at least 80% sequence identity or homology with the MIC-1 binding domain of the extracellular region of wtGFRAL, and retains at least 10% of the MIC-1 binding affinity of wtGFRAL. In a further embodiment, the GFRAL variant has at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, s at least 98%, at least 99% or 100% sequence identity with the MIC-1 binding domain of the extracellular region of wild type hGFRAL, and retains at least 10% of the MIC-1 binding affinity of wild type hGFRAL. In further embodiments, the GFRAL variant retains such as at least 20%, such as at least 40%, such as at least 60%, such as at least 80% of the MIC-1 binding affinity of wild type hGFRAL.

“GFRAL ligand binding receptor” as used herein means a receptor comprising an extracellular region, which comprises a GFRAL ligand binding segment that is derived from the GFRAL ligand binding domain in the extracellular region of GFRAL. In a further embodiment, GFRAL is derived from mammalian origin, such as human, monkey, rat, mouse, rabbit, etc. In one embodiment, the GFRAL ligand binding receptor further comprises a transmembrane region and an intracellular region. In a further embodiment, the intracellular region comprises a domain for effecting a response or change in the cytoplasm. In yet a further embodiment, the intracellular region comprises a catalytic domain. In one embodiment, a GFRAL ligand binds to the GFRAL ligand receptor in such a way that the conformation of the intracellular region of the receptor molecule changes, and such change activates or inhibits the enzymatic activity of the catalytic domain in the intracellular region. In another embodiment, a GFRAL ligand binds to the GFRAL ligand binding receptor in such a way that changes the conformation of whole receptor molecule, and such change triggers formation of a cluster/complex of GFRAL ligand, GFRAL ligand binding receptor with another receptor, so that a response or change in the cytoplasm is elicited. Preferably the other receptor is a cell surface receptor.

“MIC-1 binding receptor” as used herein means a receptor comprising an extracellular region, which comprises a MIC-1 binding segment that is derived from the MIC-1 binding domain in the extracellular region of GFRAL. In a further embodiment, GFRAL is derived from mammalian origin, such as human, monkey, rat, mouse, rabbit, etc. In one embodiment, the MIC-1 binding receptor further comprises a transmembrane region and an intracellular region. In a further embodiment, the intracellular region comprises a domain for effecting a response or change in the cytoplasm. In yet a further embodiment, the intracellular region comprises a catalytic domain. In one embodiment, a MIC-1 compound binds to the MIC-1 binding receptor in such a way that the conformation of the intracellular region of the receptor molecule changes, and such change activates or inhibits the enzymatic activity of the catalytic domain in the intracellular region. In another embodiment, a MIC-1 compound binds to the MIC-1 binding receptor in such a way that changes the conformation of whole receptor molecule, and such change triggers formation of a cluster/complex of MIC-1, MIC-1 binding receptor with another receptor, so that a response or change in the cytoplasm is elicited. Preferably the other receptor is a cell surface receptor.

In one embodiment of the invention, the “MIC-1 binding receptor comprising a MIC-1 binding segment derived from GFRAL” is a full length wild type GFRAL of mammalian origin, such as human, monkey, rat, mouse, rabbit, etc. MIC-1 binding receptor can also be allelic variations, natural mutants of GFRAL, or a protein encoded by DNA which hybridise under high or low stringency conditions to nucleic acids which encode GFRAL, etc.

In another embodiment of the invention, “MIC-1 binding receptor comprising a MIC-1 binding segment derived from GFRAL” is a GFRAL derived receptor, which is a modified GFRAL wherein one or more amino acid residues of wild type GFRAL have been substituted, deleted and/or wherein one or more natural or unnatural amino acid residues have been added, and such modified GFRAL comprises a MIC-1 binding segment derived from the MIC-1 binding domain of GFRAL and retains MIC-1 binding ability. In one embodiment, the MIC-1 binding segment of the MIC-1 binding receptor is derived from the MIC-1 binding domain of hGFRAL.

In another embodiment, MIC-1 binding receptor is a fusion protein, wherein the extracellular region comprises a MIC-1 binding segment derived from GFRAL. In one embodiment, the MIC-1 binding receptor comprises the extracellular region of GFRAL and an intracellular region of another receptor. In a further embodiment, the intracellular region comprises a catalytic domain.

In some embodiments, the MIC-1 binding domain of the extracellular region of a GFRAL derived receptor has at least 80% sequence identity or homology with the MIC-1 binding domain of the extracellular region of wtGFRAL, and retain at least 10% of the MIC-1 binding affinity of wtGFRAL. In a further embodiment, the MIC-1 binding segment has at least 85%, at least 90%, at least 91%, at least 92%, at least 93%, at least 94%, at least 95%, at least 96%, at least 97%, s at least 98%, at least 99% or 100% sequence identity with the MIC-1 binding domain of the extracellular region of wild type hGFRAL, and typically at least 10% of the MIC-1 binding affinity of wild type hGFRAL. In further embodiments, said identities to the MIC-1 binding domain of the extracellular region of wild type hGFRAL are coupled to at least 10%, such as at least 20%, such as at least 40%, such as at least 60%, such as at least 80% of the MIC-1 binding affinity of wild type hGFRAL.

The inventors of the present application surprisingly found that although GFRAL is a cell surface receptor that binds to MIC-1, in order to mediate MIC-1 activity, further binding of the complex of GFRAL and MIC-1 to RET is needed. As demonstrated in the examples, MIC-1 did not induce downstream signals in cells expressing GFRAL only or RET only, but MIC-1 signaled in cells expressing both GFRAL and RET. Without being bound to theory, it is believed that the in vivo activity of MIC-1 is mediated through both GFRAL and RET by forming a ternary complex.

RET is an abbreviation for “rearranged during transfection”, as the DNA sequence of this gene was originally found to be rearranged within a 3T3 fibroblast cell line following its transfection with DNA taken from human lymphoma cells. The natural alternative splicing of the human RET gene results in the production of 3 different isoforms of the protein RET: RET51, RET43 and RET9. These three isoforms share the same 1063 amino acids in their N-terminal, but then contain 51, 43 and 9 different amino acids in their C-terminal at the cytoplasmic side respectively. Experiment results described below show that the activity of MIC-1 is only mediated by human RET51.

Similarly for mouse, rat, and cynomolgus, there are also isoforms of RET in each species. In mouse, they were also defined as Ret9 and Ret51. Experimental results of the invention showed that the activity of MIC-1 is mediated by the mouse, rat or cynomolgus homologs of human RET51.

The term “identity” or “homology” refers to a relationship between the sequences of two or more proteins, as determined by comparing the sequences. In the art, “identity” also means the degree of sequence relatedness between proteins, as determined by the number of matches between strings of two or more amino acid residues. “Identity” measures the percent of identical matches between the smaller of two or more sequences with gap alignments (if any) addressed by a particular mathematical model or computer program (i.e., “algorithms”). Identity of related proteins is capable of being readily calculated by known methods. Such methods include, but are not limited to, those described in Computational Molecular Biology, Lesk, A. M., ed., Oxford University Press, New York, 1988; Biocomputing: Informatics and Genome Projects, Smith, D. W., ed., Academic Press, New York, 1993.

In one embodiment, results of cell-based potency assays are typically expressed as the ‘relative potency’ when compared to a reference standard or reference compound. The use of relative potency allows direct comparison between the molecules to be tested and the reference compound within the same assay, therefore reducing the impact of run-to-run variability on final reportable results.

In cell-based potency assays, a reference compound is usually used to assign relative potency, ensuring the measurement of potency is normalized over various compounds to be tested. As used herein, “reference compound” in a GFRAL ligand potency assays refers to a GFRAL ligand with known biological activity. For example, such GFRAL ligand can be a MIC-1 compound with known MIC-1 biological activity. In one embodiment of the invention, a reference compound is a wild type GFRAL ligand, for example, a wild type MIC-1 of mammalian origin, such as human, monkey, rat, mouse, etc., and a recombinant wild type MIC-1, such as recombinant human, monkey, rat, mouse MIC-1. In another embodiment of the invention, a reference compound is a variant, analogue, or derivative of the wild type GFRAL ligand with known biological activity, for example, a MIC-1 variant, analogue, or derivative, wherein the biological activity of such MIC-1 variant, analogue, or derivative is already known. In a further embodiment, the reference material is a representative batch of commercial MIC-1 compound for therapeutic use. In one embodiment of the invention, the indicator cells are grown in culture plates and stimulated with the reference compound and the GFRAL ligand to be tested respectively over a range of concentrations. In a further embodiment of the invention, the range of concentrations covers the whole dose response range from 0 to a maximal concentration. In yet a further embodiment, the whole dose response curve is in a sigmoidal shape.

In another aspect, the invention provides an assay or method for detecting the binding affinity of a GFRAL ligand. In a particular aspect, the invention provides an assay or method for detecting the binding affinity of a MIC-1 compound.

The term “binding affinity” is herein used as a measure of the strength of a noncovalent interaction between two molecules, such as a ligand (e.g., MIC-1 compound) and its receptor (MIC-1 binding receptor). Binding of a ligand (agonist or antagonist) to its receptor is the initial step in reactions that cause a biological or pharmacological effect. Binding assays is capable of being used in many ways. For example, they I used for screening of new chemical entities and also for the discovery of endogenous ligands. Such assays can also be used in a quantitative way to determine an unknown amount of analyte that is present in a test sample. They can also be used to study the receptor itself, such as identifying receptor subtypes or variants or determining the in vivo distribution of the receptor.

Binding affinity can be quantified by determination of the dissociation constant (KD). In turn, KD can be determined by measurement of the kinetics of complex formation and dissociation, e.g. by the SPR method (Biacore). The rate constants corresponding to the association and the dissociation of a monovalent complex are referred to as the association rate constants ka and dissociation rate constant kd, respectively. KD is related to ka and kd through the equation KD=ka/kd. KD is inversely proportional to the ligand's binding affinity to the receptor.

Binding affinities associated with different molecules, e.g. the binding affinities of different MIC-1 compounds, may be compared by comparison of the KD values for the individual ligand, e.g., MIC-1 compound.

Similarly, the specificity of an interaction between two molecules (e.g., a MIC-1 compound and a MIC-1 binding protein) may be assessed by determination and comparison of the KD value for the interaction of interest, e.g. a specific interaction between a MIC-1 compound and the MIC-1 binding protein, with the KD value of an interaction not of interest.

Competitive assays are widely used for binding assays, where the receptor is contacted with a labeled ligand having known binding affinity to the receptor and a test analyte whose binding affinity to the receptor is being measured or detected. The bound ligands and the free ligands are then separated to detect or measure the binding. The amount of the bound test compound is inversely proportional to the amount of bound labeled ligand.

In one embodiment, the binding assay or method comprises contacting a test sample comprising the GFRAL ligand with a GFRAL ligand binding protein, and then detecting the binding of the GFRAL ligand to the GFRAL ligand binding protein, wherein the GFRAL ligand binding protein comprises a GFRAL ligand binding segment derived from GFRAL. In another embodiment, such assay or method comprises contacting a test sample comprising a reference compound and the GFRAL ligand to be tested with a GFRAL ligand binding protein, then detecting the binding of the reference compound to the GFRAL ligand binding protein, wherein the reference compound is a different GFRAL ligand with known binding affinity and is labeled for detection. In a further embodiment, the relative binding affinity of the GFRAL ligand is calculated based on the binding of the reference compound.

In a further embodiment, the binding assay or method comprises contacting a test sample comprising the MIC-1 compound with a MIC-1 binding protein, and then detecting the binding of the MIC-1 compound to the MIC-1 binding protein, wherein the MIC-1 binding protein comprising a MIC-1 binding segment derived from GFRAL. In another embodiment, such assay or method comprises contacting a test sample comprising a reference compound and the MIC-1 compound to be tested with a MIC-1 binding protein, then detecting the binding of the reference compound to the MIC-1 binding protein, wherein the reference compound is a different MIC-1 compound with known binding affinity and is labeled for detection. In a further embodiment, the relative binding affinity of the MIC-1 compound is calculated based on the binding of the reference compound.

“GFRAL ligand binding protein comprising GFRAL ligand binding segment derived from GFRAL” can be any form of protein that comprises a GFRAL ligand binding segment derived from GFRAL. In one embodiment, it could a GFRAL receptor. In another embodiment, the GFRAL ligand binding protein comprises a tag, e.g., HPC4, Fc, Fab, polyhistidine-tag, antibody light chain, etc. In one embodiment, the GFRAL ligand binding protein is soluble. In another embodiment, the GFRAL ligand binding protein is anchored on a solid phase or a cell surface. In a further embodiment, the solid phase is a plate or a bead. In a further embodiment, the cell is immobilized. In one embodiment, the GFRAL ligand binding protein comprises the extracellular region of wild type hGFRAL or mGFRAL. In one embodiment, the GFRAL ligand binding protein comprises a reporter peptide, except for the GFRAL ligand binding segment derived from GFRAL. In a further embodiment, the reporter peptide is luciferase, bacterial β-galactosidase, trpE, Protein A, β-lactamase, alpha amylase, alcohol dehydrogenase, yeast alpha mating factor, etc.

“MIC-1 binding protein comprising MIC-1 binding segment derived from GFRAL” can be any form of protein that comprises a MIC-1 binding segment derived from GFRAL. In one embodiment, it could a MIC-1 receptor. In another embodiment, the MIC-1 binding protein comprises a tag, e.g., HPC4, Fc, Fab, Fab-HPC4, polyhistidine-tag, antibody light chain, etc. In one embodiment, the MIC-1 binding protein is soluble. In another embodiment, the MIC-1 binding protein is anchored on a solid phase or a cell surface. In a further embodiment, the solid phase is a plate or a bead. In a further embodiment, the cell is immobilized. In one embodiment, the MIC-1 binding protein comprises the extracellular region of wild type hGFRAL or mGFRAL. In one embodiment, the MIC-1 binding protein comprises a reporter peptide, except for the MIC-1 binding segment derived from GFRAL. In a further embodiment, the reporter peptide is luciferase, bacterial β-galactosidase, trpE, Protein A, β-lactamase, alpha amylase, alcohol dehydrogenase, yeast alpha mating factor, etc.

In GFRAL ligand binding assays, “reference compound” refers to a GFRAL ligand with known binding affinity to GFRAL ligand binding receptor. In one embodiment of the invention, the reference compound used in binding assay is a wild type GFRAL ligand of mammalian origin, such as human, monkey, rat, mouse, etc., and a recombinant wild type GFRAL ligand, such as recombinant human, monkey, rat, mouse GFRAL ligand. In another embodiment of the invention, a reference compound used in binding assay is a GFRAL ligand variant, analogue, or derivative, wherein the binding affinity of such GFRAL ligand variant, analogue, or derivative is already known.

In an embodiment, the GFRAL ligand is MIC-1 compound. In MIC-1 compound binding assays, “reference compound” refers to a MIC-1 compound with known binding affinity to MIC-1 binding receptor. In one embodiment of the invention, the reference compound used in binding assay is a wild type MIC-1 of mammalian origin, such as human, monkey, rat, mouse, etc., and a recombinant wild type MIC-1, such as recombinant human, monkey, rat, mouse MIC-1. In another embodiment of the invention, a reference compound used in binding assay is a MIC-1 variant, analogue, or derivative, wherein the binding affinity of such MIC-1 variant, analogue, or derivative is already known.

Labels are widely used in binding assays. In one embodiment, the labels can be detected directly. For example, such directly detectable label a radio-isotopic label, a label detected by colorimetric detection system, a label detected by fluorescence detection system, a label detected by luminescence detection system, etc. In a further embodiment, the detectable label is 3H, 125I or 32P. In another further embodiment, the detectable label is tetramethylbenzidine (TMB), fluorescein, Alexa Fluor dyes, Bodipy FL-propionic acid, methoxycoumarin-COOH, CyDyes, Dansyl-SE, Fluorescein, NBD-SE, Rhodamine Green-SE, Texas Red-SE, Europium-chloride.6H2O, luminol, lucigenin, luciferin, etc.

In another embodiment, the labels cannot be detected directly, but can facilitate binding of the labelled ligand to a binding partner that is labeled with a directly detectable label. In a further embodiment, such binding facilitating label is biotin, digoxin, HPC4, etc.

Ligand-receptor binding can be detected by many technologies. Some technologies require using a label. Some technologies are label-free. For example, binding can be detected by SPA (scintillation proximity assay), TRF (time-resolved fluorescence), FRET (fluorescence resonance energy transfer), BRET (bioluminescence resonance energy transfer), TR-FRET (time-resolved fluorescence resonance energy transfer), FP (fluorescence polarization), FMAT (fluorometric microvolume assay technology), AlphaScreen™, flow cytometry, FCS (fluorescence correlation spectroscopy), SPR (surface plasmon resonance), ForteBio, or TIRF (total internal reflection fluorescence), etc. Please see L. A. A. deJong et al., Receptor-ligand binding assays: Technologies and Applications, J. Chromatogr. B 829 (2005) 1-25.

The foregoing descriptions and the following list of embodiments are non-limiting to the invention:

Embodiment 1: A method for determining the activity of a GFRAL ligand, such as a MIC-1 compound, comprising:

(a) contacting an indicator cell with a test sample that comprises the GFRAL ligand, such as a MIC-1 compound; and

(b) detecting a biological response of the indicator cell contacted with the test sample;

wherein the indicator cell expresses on the cell surface a MIC-1 binding receptor comprising the MIC-1 binding segment derived from GFRAL.

Embodiment 2: The method of embodiment 1, wherein the GFRAL ligand is a MIC-1 compound, and wherein the indicator cell further expresses a cell surface receptor kinase, and the biological response is induced when the MIC-1 compound, the MIC-1 compound binding receptor, and the cell surface receptor kinase form a ternary complex.

Embodiment 3: The method of embodiment 2, wherein the cell surface receptor kinase is RET receptor tyrosine kinase.

Embodiment 4: The method according to any one of embodiments 1-3, wherein GFRAL is derived from human, mouse, rat, rabbit or cynomolgus.

Embodiment 5: The method according to any one of embodiments 1-4, wherein the indicator cell also expresses a reporter peptide, such as luciferase, bacterial β-galactosidase, trpE, Protein A, β-lactamase, alpha amylase, alcohol dehydrogenase, yeast alpha mating factor, etc.

Embodiment 6: The method of according to any one of embodiments 1-5, wherein the indicator cell exogenously expresses the MIC-1 binding receptor, and/or the cell surface receptor kinase.

Embodiment 7: The method of embodiment 6, wherein the indicator cell is a mammalian cell.

Embodiment 8: The method of embodiment 7, wherein the indicator cell is BHK21 cell, PC-12 cell, HEK293 cell, or HEK293 cell stably expressing the tetracycline (Tet) repressor.

Embodiment 9: The method according to any one of embodiments 1-8, wherein the MIC-1 binding receptor is full length GFRAL.

Embodiment 10: The method according to any one of embodiments 1-8, wherein the MIC-1 binding receptor comprises the extracellular region of GFRAL.

Embodiment 11: The method of embodiment 1 or 2, wherein the biological response is related to signal transduction.

Embodiment 12: The method of embodiment 11, wherein the signal transduction related biological response is increase or decrease of a protein kinase phosphorylation, transcription of a gene or expression of a protein.

Embodiment 13: The method of embodiment 12, wherein the protein kinase is the cell surface receptor kinase or an intracellular protein kinase.

Embodiment 14: The method of embodiment 13, wherein the signal transduction related biological response is related to activation of Hedgehog pathway, MEF2 pathway, Smad pathway, ERK/MAPK pathway, JNK/p38 pathway, Small GTPase pathway, PI3K/Akt pathway, PLC-γ pathway, JAK/STAT pathway, Src-family kinases, CDK5 (cyclin-dependent kinase 5), Src-like kinase Fyn, FAK (focal adhesion kinase), and a combination thereof.

Embodiment 15: The method of embodiment 14, wherein the signal transduction related biological response is increased or decreased phosphorylation of a protein kinase of ERK/MAPK pathway.

Embodiment 16: The method of embodiment 15, wherein the protein kinase is selected from a group consisting of ERK1/2, MAP3K, Ras, MEK, Shc, c-Raf, or a combination thereof. Embodiment 17: The method of embodiment 14, wherein the signal transduction related biological response is increased or decreased phosphorylation of a protein kinase of PI3K/Akt pathway.

Embodiment 18: The method of embodiment 17, wherein the protein kinase is selected from the group consisting of PI3K (phosphoinositide 3-kinase), Akt1/2/3, mTOR (mammalian target of rapamycin), 4E-BP1 (4E-binding protein 1), S6 kinase, c-Ab1, and a combination thereof.

Embodiment 19: The method of embodiment 14, the signal transduction related biological response is increased or decreased phosphorylation of a protein kinase of Smad pathway, wherein the protein kinase could be selected from the group consisting of Smad1, Smad2, Smad3, Smad5, Smad8, and a combination thereof.

Embodiment 20: The method of embodiment 14, wherein the signal transduction related biological response is increased or decreased phosphorylation of a protein kinase of JNK/p38 MAPK pathway, wherein the protein kinase could be selected from a group consisting of MKK3, MKK4, MKK6, JNK, p38 MAPK, TAK1, TRAF6, and a combination thereof.

Embodiment 21: The method of embodiment 14, wherein the signal transduction related biological response is increased or decreased phosphorylation of a protein kinase of Small GTPase pathway, wherein the protein kinase could be selected from the group consisting of RhoA, Rac, Cdc42, and a combination thereof.

Embodiment 22: The method of embodiment 14, wherein the signal transduction related biological response is increased or decreased phosphorylation of a protein kinase of JAK/STAT pathway, wherein the protein kinase is selected from the group consisting of JAK1, JAK2, JAK3, TYK2, STAT1, STAT2, STAT3, STATS and a combination thereof. Embodiment 23: The method of embodiment 14, wherein the signal transduction related biological response is up-regulated or down-regulated transcription of a gene or expression a protein of Hedgehog pathway, wherein the gene or protein could be GLI.

Embodiment 24: The method of embodiment 14, wherein the signal transduction related biological response is up-regulated or down-regulated transcription or a gene or expression of a protein of MEF2 pathway, wherein the gene or protein could be MEF2.

Embodiment 25: The method of embodiment 21, wherein the increase or decrease of phosphorylation is determined by the phosphorylation level of the protein kinase; or is determined by the activity level of the reporter peptide, wherein the activity level of the reporter peptide is associated with the phosphorylation level of the protein kinase.

Embodiment 26: The method of embodiment 25, wherein the reporter peptide is luciferase.

Embodiment 27: The method of embodiment 25, wherein the phosphorylation of a protein kinase is measured by a process comprising:

(a) lysing the indicator cell to obtain cell lysate;

(b) exposing the cell lysate to a capture agent that specifically binds the protein kinase;

(c) removing the unbound cell lysate;

(d) exposing the bound protein kinase to a phosphor-specific antibody against a phosphorylated amino acid; and

(e) detecting binding of the phospho-specific antibody.

Embodiment 28: The method of embodiment 27, wherein the phospho-specific antibody is an anti-phosphotyrosine antibody, an anti-phosphoserine antibody, or an antiphosphotyrosine antibody.

Embodiment 29: The method of embodiment 25, wherein the phosphorylation of a protein kinase is measured by a process comprising:

(a) lysing the indicator cell to obtain cell lysate;

(b) exposing the cell lysate to a phospho-specific antibody against the phosphorylated protein kinase; and

(c) detecting binding of the phospho-specific antibody.

Embodiment 30: The method of embodiment 11, wherein the signal transduction related biological response is increase or decrease of the level of a second messenger.

Embodiment 31: The method of embodiment 30, wherein the second messenger is selected from the group consisting of diacylglycerol, phosphatidylinositol, cAMP, cGMP, IP3, Ca2+, nitric oxide (NO), carbon monoxide (CO), hydrogen sulfide (H2S), and a combination thereof.

Embodiment 32: The method of embodiment 1, wherein the indicator cell endogenously expresses the MIC-1 binding receptor, and/or the cell surface receptor kinase; furthermore, the indicator cell is derived from neuron cells.

Embodiment 33: The method of embodiment 32, wherein the neuron cell is derived from hypothalamus or brain stem.

Embodiment 34: The method of embodiment 33, wherein the neuron cell is derived from arcuate nucleus (ARC), paraventricular nucleus (PVN), area postrema (AP), or nucleus tractus solitarus (NTS).

Embodiment 35: The method of embodiment 1, wherein the MIC-1 binding receptor is a fused protein comprising the extracellular region of GFRAL, a transmembrane domain, and an intracellular region of a cell surface receptor.

Embodiment 36: The method of embodiment 35, wherein the intracellular region comprises a catalytic domain, and the biological response is the increase or decrease of the catalytic activity of the catalytic domain.

Embodiment 37: The method of embodiment 35, wherein the cell surface receptor kinase is a member receptor of TGF-β super family.

Embodiment 38: The method of embodiment 1, wherein the biological response is related to cell proliferation or cell validity.

Embodiment 39: The method of embodiment 1, wherein the biological response is related to cell apoptosis or cell growth arrest.

Embodiment 40: The method of embodiment 39, wherein the biological response is caspase activity.

Embodiment 41: The method of embodiment 39, wherein the biological response is measured as the number of viable cells or the number of apoptotic cells.

Embodiment 42: The method of embodiment 1, wherein the biological response is related to cytotoxicity.

Embodiment 43: The method of embodiment 1, wherein the biological response is up-regulated transcription of a gene.

Embodiment 44: The method of embodiment 43, wherein the gene is selected from a group consisting of GLI, MEF2, SRE, c-fos, POMC (pro-opiomelanocortin), CART (cocaine and amphetamine regulated transcript), or a combination thereof.

Embodiment 45: The method of embodiment 1, wherein the biological response is down-regulating mRNA of a gene.

Embodiment 46: The method of embodiment 45, wherein the gene is NPY (neuropeptide Y), AGRP (agouti-related protein), or a combination thereof.

Embodiment 47: A cell line for determining the activity of a MIC-1 compound, wherein the cell line expresses a MIC-1 binding receptor and a cell surface receptor kinase, wherein the MIC-1 binding receptor comprises the MIC-1 binding segment derived from GFRAL, and a biological response is induced when the MIC-1 compound, the MIC-1 binding receptor, and the cell surface receptor kinase form a ternary complex.

Embodiment 48: The cell line of embodiment 47, wherein the cell surface receptor kinase is RET receptor tyrosine kinase.

Embodiment 49: A cell line for determining the activity of a GFRAL ligand, wherein the cell lines expresses a GFRAL ligand binding receptor and RET receptor tyrosine kinase, wherein the GFRAL ligand binding receptor comprises the GFRAL ligand binding segment derived from GFRAL, and a biological response is induced when the GFRAL ligand, the GFRAL ligand binding receptor, and RET form a ternary complex.

Embodiment 50: The cell line of embodiment 47 or 49, wherein the cell line also expresses a reporter peptide, and the biological response is capable of being detected by detecting the activity of the reporter peptide.

Embodiment 51: The cell line of embodiment 50, wherein the reporter peptide is luciferase, bacterial β-galactosidase, trpE, Protein A, β-lactamase, alpha amylase, alcohol dehydrogenase, yeast alpha mating factor, etc.

Embodiment 52: The cell line of embodiment 51, wherein the reporter peptide is luciferase.

Embodiment 53: The cell line of embodiment 52, wherein the cell line also expresses SRE.

Embodiment 54: The cell line of embodiment 47 or 49, wherein the cell line is derived a mammalian cell, such as BHK21 cell, HEK293 cell, or PC-12 cell.

Embodiment 55: The cell line of embodiment 47 or 49, wherein the cell line expresses full length hGFRAL, full length hRET, and luciferate.

Embodiment 56: A method for detecting the binding affinity of a GFRAL ligand, such as a MIC-1 compound and GFRAL, comprising:

(a) contacting a test sample comprising the GFRAL ligand, such as a MIC-1 compound with a MIC-1 binding protein; and

(b) detecting binding of between the GFRAL ligand, such as a MIC-1 compound and the MIC-1 binding protein,

wherein the MIC-1 binding protein comprising the MIC-1 binding segment derived from GFRAL.

Embodiment 57: A method for detecting the binding affinity of a MIC-1 compound, comprising:

(a) contacting a test sample with a MIC-1 binding protein, wherein the test sample comprises a reference compound; and

(b) detecting binding of the reference compound to the MIC-1 binding protein;

wherein

the reference compound is a different MIC-1 compound with known binding affinity, and,

the MIC-1 binding protein comprising the MIC-1 binding segment derived from GFRAL.

Embodiment 58: The method of embodiment 55, wherein the MIC-1 binding segment comprises the extracellular region of GFRAL.

Embodiment 59: The method of embodiment 55, wherein the MIC-1 binding protein comprises C1 and C2 domains of GFRAL.

Embodiment 60: The method of embodiment 55, wherein the MIC-1 binding protein is anchored on a cell surface.

Embodiment 61: The method of embodiment 55, wherein GFRAL is derived from human, mouse, rat, rabbit or cynomolgus.

Embodiment 62: The method of embodiment 53, wherein the MIC-1 binding protein is immobilized on a solid phase.

Embodiment 63: The method of embodiment 56, wherein the reference compound is immobilized.

Embodiment 64: The method of embodiment 56, wherein reference compound is labeled for detection.

Embodiment 65: The method of embodiment 63, wherein the label for detection is a radio-isotope, biotin, digoxin, tetramethylbenzidine (TMB), fluorescein, luminol, lucigenin, or luciferin.

Embodiment 66: The method of embodiment 55, wherein the binding is detected by radioactivity detection system, colour detection systems, fluorescence detection systems, or luminescence detection systems.

Embodiment 67: The method of embodiment 55, wherein binding is detected by SPA (scintillation proximity assay), TRF (time-resolved fluorescence), FRET (fluorescence resonance energy transfer), BRET (bioluminescence resonance energy transfer), TR-FRET (time-resolved fluorescence resonance energy transfer), FP (fluorescence polarization), FMAT (fluorometric microvolume assay technology), AlphaScreen™, flow cytometry, FCS (fluorescence correlation spectroscopy), SPR (surface plasmon resonance), or TIRF (total internal reflection fluorescence).

Embodiment 68: A method for determining the activity of a GFRAL ligand, comprising:

(a) contacting an indicator cell with the GFRAL ligand, and detecting a biological response of the indicator cell contacted with GFRAL ligand;

(c) contacting the indicator cell with a MIC-1 compound with known activity, and detecting a biological response of the indicator cell contacted with the MIC-1 compound;

(c) comparing the biological response induced by the GFRAL ligand with the biological response induced by the MIC-1 compound,

wherein the indicator cell expresses on the cell surface a MIC-1 binding receptor comprising the MIC-1 binding segment derived from GFRAL.

Embodiment 69: A method for determining the activity of a GFRAL variant, comprising:

(a) contacting an indicator cell with a test sample that comprises the GFRAL variant and a MIC-1 compound with known activity; and

(b) detecting a biological response of the indicator cell contacted with the test sample;

wherein the indicator cell expresses on the cell surface a MIC-1 binding receptor comprising the MIC-1 binding segment derived from GFRAL.

All references, including publications, patent applications, and patents, cited herein are hereby incorporated by reference in their entirety and to the same extent as if each reference were individually and specifically indicated to be incorporated by reference and were set forth in its entirety herein (to the maximum extent permitted by law).

All headings and sub-headings are used herein for convenience only and should not be construed as limiting the invention in any way.

The use of any and all examples, or exemplary language (e.g., “such as”) provided herein, is intended merely to better illuminate the invention and does not pose a limitation on the scope of the invention unless otherwise claimed. No language in the specification should be construed as indicating any non-claimed element as essential to the practice of the invention.

The citation and incorporation of patent documents herein is done for convenience only and does not reflect any view of the validity, patentability, and/or enforceability of such patent documents.

This invention includes all modifications and equivalents of the subject matter recited in the claims appended hereto as permitted by applicable law.

EXAMPLES Example 1 Generation of Human and Mouse GFRAL Extracellular Region Expression Constructs Reagents: Qiagen EndoFree Plasmid Maxi Kit (#12362), Gene Synthesis and Cloning of Expression Constructs

hGFRAL ECR-pCLC

Human (Homo sapiens) GDNF family receptor alpha like (hGFRAL) DNA sequence (SEQ ID: 1) is from CCDS4957.1. The sequence was analyzed with Phobius, SignalP and TMHMM. The definition of its extracellular region (ECR) is consistent with the UniProt record Q6UXV0. Only the sequence encoding the ECR (amino acids 19-351 in SEQ ID: 1) was retained while the sequences encoding the signal peptide, transmembrane, and intracellular regions were discarded. The DNA sequence encoding hGFRAL_ECR was further optimized to remove common restriction sites (EcoRI, BamHI, HindIII, and ApaI) while retaining the encoded ECR peptide sequence.

hGFRAL_ECR DNA was synthesized and cloned into expression vector pCLC. This expression vector contains a CMV promoter to drive expression in the mammalian cells. It has DNA sequence encoding CD33 signal peptide for protein secretion. It has DNA sequence encoding mouse anti-TNP light chain (ATNP LC or LC) sequence. This construct, when co-expressing in a cell with plasmid encoding a peptide comprising the anti-TNP fab heavy chain (HC) and a fragment of human PDGFRB containing the transmembrane region (HC_TM, the amino sequence is shown in SEQ ID NO: 14), would create anti-TNP Fab protein and anchored the hGFRAL_ECR on the cell surface through the PDGFRB transmembrane region. The hGFRAL_ECR DNA was cloned in frame directly after the sequence of CD33 signal peptide and in front of the ATNP LC sequence. A short linker sequence encoding GGGGSGGGGS peptide was also inserted between the hGFRAL_ECR and ATNP LC sequences to provide flexibility between the fused domains. The name of the expression plasmid is hGFRAL_ECR-pCLC. The amino acid sequence of the protein expressed by hGFRAL_ECR-pCLC is shown in SEQ ID NO: 6.

Plasmids:

hGFRAL_C1-pCLC,

hGFRAL_C2-pCLC,

hGFRAL_C3-pCLC,

hGFRAL_C1C2-pCLC, and

hGFRAL_C2C3-pCLC

For the mapping of the ligand binding domain in the ECR of hGFRAL, five expression constructs were generated encoding different domains of hGFRAL. Based on homology and structural predictions, there are three separate cysteine rich domains in the hGFRAL_ECR. They are amino acid 19-130 (referred as C1 from here on), amino acid 131-219 (referred as C2 from here on), and amino acid 220-351 (referred as C3 from here on). The amino acids refer to the peptide sequence in UniProt record Q6UXV0. We generated the expression constructs encoding these domains: C1, C2, C3, C1C2, and C2C3. These constructs are in the same expression format as the full length hGFRAL_ECR-LC construct described as above. They contain the N-terminal CD33 signal peptide sequence and the C terminal ATNP LC sequence. The names of these expression plasmids are hGFRAL_C1-pCLC, hGFRAL_C2-pCLC, hGFRAL_C3-pCLC, hGFRAL_C1C2-pCLC, and hGFRAL_C2C3-pCLC. The amino acid sequences of the proteins expressed by these plasmids are shown in SEQ ID NOs: 9, 10, 11, 12, and 13 respectively.

mGFRAL_ECR-pCLC

There are two splice variants of Mouse (Mus musculus) GDNF family receptor alpha like (mGFRAL), the full-length cell membrane-anchored form, isoform 1 (DNA sequence is shown in SEQ ID NO: 3; amino acid sequence is shown in SEQ IN NO: 4); and a short secreted form, isoform 2, which only consisting of the signal peptide and the ECR as of isoform 1. The DNA sequence of mGFRAL isoforms 1 is from CCDS40694.1. The definition of its extracellular region (referred as mGFRAL_ECR from here on) is based on UniProt record Q63JE0-1. Only the sequence encoding the ECR was retained in the construct while the sequences predicted to encode the signal peptide, transmembrane, and intracellular regions were discarded. The DNA sequence encoding mGFRAL_ECR was further optimized to remove common restriction sites (EcoRI, BamHI, HindIII, and ApaI) while retaining the encoded ECR peptide sequence.

mGFRAL_ECR DNA was synthesized and cloned into expression vector pCLC. This expression vector contains a CMV promoter to drive expression in the mammalian cells. It has DNA sequence encoding CD33 signal peptide for protein secretion. It has DNA sequence encoding mouse anti-TNP light chain (ATNP LC or LC) sequence. This construct, when co-expressing with plasmid encoding a peptide comprising the anti-TNP fab heavy chain (HC) and a fragment of human PDGFRB containing the transmembrane region (HC_TM, the amino sequence is shown in SEQ ID NO: 14), would create anti-TNP Fab protein and anchored the mGFRAL_ECR on the cell surface through the PDGFRB transmembrane region. The mGFRAL_ECR DNA was cloned in frame directly after the CD33 signal sequence and in front of the ATNP LC sequence. A short linker sequence encoding GGGGSGGGGS peptide was also inserted between the mGFRAL_ECR and ATNP LC sequences to provide flexibility between the fused domains. The name of the expression plasmid is mGFRAL_ECR-pCLC. The amino acids sequence of the protein expressed by mGFRAL_ECR-pCLC is shown in SEQ ID NO: 8.

pJSV002-ATNP-mIgG1-HL-HPC4

The coding sequence of the heavy chain of an ATNP Fab fused with HPC4 at its C terminus was synthesized and cloned into expression vector pJSV002 (HC_HPC4, the amino sequence is shown in SEQ ID NO: 15). This expression vector contains a CMV promoter to drive expression in the mammalian cells. It has DNA sequence encoding CD33 signal peptide for protein secretion. When the construct is co-transfected into mammalian cells with plasmid encoding the ATNP LC or a fusion protein comprising ATNP LC, the ATNP Fab or a fusion protein comprising ATNP Fab will be formed, e.g., hGFRAL(ECR)-Fab, mGFRAL(ECR)-Fab.

pJSV002-CD33-hGFRAL ECR-hIgG1.1 Fc

hGFRAL_ECR DNA was synthesized and cloned into expression vector pJSV002-CD33-hIgG1.1_Fc. This vector has the same CMV promoter to drive expression. The hGFRAL_ECR was cloned in frame between the CD33 signal peptide and the hIgG1.1_Fc sequence. The amino acid sequence of the fusion protein is hGFRAL_ECR-hIgG1.1_Fc (SEQ ID NO: 7, also “hGFRAL-Fc”). The name of the expression plasmid is pJSV002-CD33-hGFRAL_ECR-hIgG1.1_Fc.

Example 2 Expression of Soluble Form of Human and Mouse GFRAL Extracellular Region Fusion Proteins, hGFRAL(ECR)-Fab, hGFRAL(ECR)-Fc, and mGFRAL(ECR)-Fab Cell: HEK293-6E Reagents:

Gibco® FreeStyle™ 293 Expression Medium, Invitrogen, Cat. No.: 12338-026;

Gibco® 10% Pluronic F68, Invitrogen, Cat. No.: 24040-032;

Gibco® Geneticin, Invitrogen, Cat. No.: 11811-023;

Gibco® Opti-MEM® I+GlutaMax™ I, Invitrogen, Cat. No.: 51985-034;

293Fectin™ Reagent, Invitrogen, Cat. No.: 12347-019;

Plasmids:

hGFRAL_ECR-pCLC

pJSV002-CD33-hGFRAL_ECR-hIgG1.1_Fc

mGFRAL_ECR-pCLC

hGFRAL_C1-pCLC

hGFRAL_C2-pCLC

hGFRAL_C3-pCLC

hGFRAL_C1C2-pCLC

hGFRAL_C2C3-pCLC

pJSV002-ATNP-mIgG1-HP-HPC4 (for TM1, type I cell surface receptor)

Transient Transfection

hGFRAL(ECR)-Fab-HPC4 (hGFRAL(ECR)-Fab), hGFRAL-hIgG1.1_Fc (hGFRAL(ECR)-Fc) and mGFRAL(ECR)-Fab-HPC4 (mGFRAL(ECR)-Fab) were produced using transient transfection in HEK293-6E cells.

HEK293-6E cells were grown in suspension in Gibco® FreeStyle™ 293 Expression Medium supplemented with 0.1% Fluronic F68 at 37° C. with 5% CO2. Cells were transfected when reaching 1×106 cells/ml density.

For 300 ml cell volume, 300 μg of DNA were used for transfection. For the two pCLC constructs, 300 μg of DNA included 240 μg of the plasmids and 60 μg pJSV002-ATNP-mIgG1-HP-HPC4.

The 300 μg of DNA for transfection was diluted in 15 ml of Opti-MEM. 300 μl of 293Fectin was added to a separate 15 ml of Opti-MEM and was gently mixed. The transfection agent was incubated for 5 minutes at room temperature. After incubation, the 15 ml diluted DNA for transfection was added to the 293Fectin mixture for a total volume of 30 ml. This mixture was then incubated for 25 minutes at room temperature to allow 293Fectin-DNA complexes to form.

After incubation, the mixture containing 293Fectin-DNA complexes was added to 300 ml of HEK293-6E cells (at 1×106 cells/ml density) in 1 L flask. The cells were then incubated in 37° C. shaking incubator with 5% CO2.

The cells were harvested 5 days post transfection. Cells were removed by centrifugation at 6000 rpm for 15 minutes followed by filtration of the supernatant using a 0.45 μm filter. The supernatant were checked for expression using SDS-PAGE gel and delivered for subsequent purification.

Example 3 Cloning and Expression of Soluble Forms of Human RET Extracellular Region, hRET(ECR)-Fc and hRET(ECR)-Fab Reagents

Qiagen EndoFree Plasmid Maxi Kit (#12362)

Gibco® FreeStyle™ 293 Expression Medium, Invitrogen, Cat. No.: 12338-026;

Gibco® 10% Pluronic F68, Invitrogen, Cat. No.: 24040-032;

Gibco® Geneticin, Invitrogen, Cat. No.: 11811-023;

Gibco® Opti-MEM® I+GlutaMax™-I, Invitrogen, Cat. No.: 51985-034;

293Fectin™ Reagent, Invitrogen, Cat. No.: 12347-019;

Cell: HEK293-6E Gene Synthesis and Cloning of Expression Constructs

Human (Homo sapiens) proto-oncogene tyrosine-protein kinase receptor's (RET) DNA sequence is from CCDS7200.1 and UniProt P07949-1 (hRET51, amino acid sequence is shown in SEQ ID NO: 5). The sequence was analyzed with Phobius, SignalP and TMHMM. The definition of its extracellular region (ECR) is consistent with the UniPort record P07949-1. Only the sequence encoding the ECR (amino acids 29-636) was retained while the sequences encoding the signal peptide, transmembrane, and intracellular regions were discarded. The DNA sequence encoding hRET_ECR was further optimized to remove common restriction sites (EcoRI, BamHI, HindIII, and ApaI) while retaining the encoded ECR peptide sequence. There is an additional splice variant of hRET with corresponding CCDS (CCDS53525.1) and UniPort P07949-2. The differences between the two splice forms are in the C terminal, which are within intracellular regions. The hRET_ECR sequences of the two splice forms are the same.

hRET_ECR DNA was synthesized and cloned into two expression vectors. The first vector was pCLC. This expression vector contains a CMV promoter to drive expression in the mammalian cell lines. It has DNA sequence encoding CD33 signal peptide for protein secretion. It has DNA sequence encoding mouse anti-TNP light chain (ATNP LC, or LC) sequence. This construct, when co-expressing with plasmid encoding the anti-TNP fab heavy chain, would create anti-TNP Fab protein. The hRET_ECR DNA was cloned in frame directly after the CD33 signal sequence and in front of the ATNP LC sequence. A short linker sequence encoding GGGGSGGGGS peptide was also inserted between the hRET_ECR and ATNP LC sequences to provide flexibility between the fused ECR and ATNP LC domains. The resulting fusion protein is hRET_ECR-LC. The name of the expression plasmid is hRET_ECR-pCLC. The second vector is pJSV002-CD33-hIgG1.1_Fc. It has the same CMV promoter to drive expression. The hRET_ECR was cloned in frame between the CD33 signal peptide and the hIgG1.1_Fc sequence. The sequence of the fusion protein is hRET_ECR-hIgG1.1_Fc. The name of the expression plasmid is pJSV002-CD33-hRET_ECR-hIgG1.1_Fc.

Plasmids for Transfection

hRET_ECR-pCLC

pJSV002-CD33-hRET_ECR-hIgG1.1_Fc

pJSV002-aTNP-mIgG1-HL-HPC4 (for TM1)

All expression plasmids were sequence verified and prepared in large quantity using Qiagen EndoFree Plasmid Maxi Kit following manufacturer's instruction.

Transient Transfection

2 Proteins were produced using transient transfection in HEK293-6E cells. They are hRET(ECR)-Fab-HPC4 (hRET(ECR)-Fab), AND hRET(ECR)-hIgG1.1_Fc (hRET(ECR)-Fc).

HEK293-6E were grown in suspension in Gibco® FreeStyle™ 293 Expression Medium supplemented with 0.1% Fluronic F68 at 37° C. with 5% CO2. Cells were transfected when reaching 1×106 cells/ml density.

For 300 ml cell volume, 300 μg of DNA were used for transfection. For the two pCLC constructs, the 300 μg of DNA included 240 μg of the plasmids and 60 μg pJSV002-aTNP-mIgG1-HL-HPC4 plasmid. For the two hIgG1.1_Fc constructs, 300 μg of plasmids DNA were used.

DNA was diluted in 15 ml of Opti-MEM. 300 μl of 293Fectin added to a separate 15 ml of Opti-MEM and was gently mixed. The transfection agent was incubated for 5 minutes at room temperature. After incubation, the 15 ml diluted DNA was added to the 293Fectin mixture for a total volume of 30 ml. This mixture was then incubated for 25 minutes at room temperature to allow 293Fectin-DNA complexes to form.

After incubation, the mixture containing 293Fectin-DNA complexes was added to 300 ml of HEK293-6E cells (at 1×106 cells/ml density) in 1 L flask. The cells were then incubated in 37° C. shaking incubator with 5% CO2.

The cells were harvested at 5 days post transfection. Cells were removed by centrifugation at 6000 rpm for 15 minutes followed by filtration of the supernatant using a 0.45 μm filter. The supernatant were checked for expression using SDS-PAGE gel and delivered for subsequent purification.

Example 4 Purification of Human and Mouse GFRAL Extracellular Regions Fusion Proteins

Purification of hGFRAL(ECR)-Fab-HPC4

The culture supernatant containing secreted hGFRAL(ECR)-Fab-HPC4 was applied to an anti-HPC4 sepharose 4FF affinity column (anti-HPC4 antibody coupled to the CNBr activated sepharose 4FF resin, 15 mL), equilibrated in 20 mM Tris-HCl, 100 mM NaCl, 1 mM CaCl2, pH 7.4. The bound hGFRAL(ECR)-Fab-HPC4 protein was eluted with 20 mM Tris-HCl, 100 mM NaCl, pH 7.4, 1 mM EGTA. Fractions were pooled and concentrated to a final volume of approx. 3.0 mL using Millipore Amicon Ultra Centrifugal Filters (UFC 901096 10K NMWL, Billerica, Mass., USA) and further purified by size-exclusion chromatography on a Hi-Load 16/60 Superdex 200 μg column (28-9893-35 GE Healthcare, Uppsala, Sweden) in phosphate buffered saline (PBS). The fractions containing monomeric hGFRAL(ECR)-Fab-HPC4 were pooled and diluted 10 fold into 20 mM sodium acetate pH5.0 and then applied to a Mono S 5/52 GL column (17-5168-01 GE Healthcare, Uppsala, Sweden). The bound hGFRAL(ECR)-Fab-HPC4 was then eluted with a 10%-50% linear gradient of 20 mM sodium acetate pH5.0, 1 M NaCl in 30 column volumes. The fractions of hGFRAL(ECR)-Fab-HPC4 were buffer-exchanged to PBS by Millipore Amicon Ultra Centrifugal Filters (UFC 901096 10K NMWL, Billerica, Mass., USA). Purified proteins were sterilized by filtration through a 0.2 m filter unit (16541 Sartorius, Goettingen, Germany). The final protein concentrations were determined by measuring 280 nm absorbance with a NANODROP UV spectrometer.

Purification of mGFRAL(ECR)-Fab-HPC4

The culture supernatant containing secreted mGFRAL(ECR)-Fab-HPC4 was applied to an anti-HPC4 sepharose 4FF affinity column (anti-HPC4 antibody coupled to the CNBr activated sepharose 4FF resin, 5 mL), equilibrated in 20 mM Tris-HCl, 100 mM NaCl, 1 mM CaCl2, pH 7.4. The bound mGFRAL(ECR)-Fab-HPC4 protein was eluted with 20 mM Tris-HCl, 100 mM NaCl, pH 7.4, 1 mM EGTA. Fractions were pooled and concentrated to a final volume of approx. 3.0 mL using Millipore Amicon Ultra Centrifugal Filters (UFC 901096 10K NMWL, Billerica, Mass., USA) and further purified by size-exclusion chromatography on a Hi-Load 16/60 Superdex 200 μg column (28-9893-35 GE Healthcare, Uppsala, Sweden) in phosphate buffered saline (PBS). The fractions containing monomeric mGFRAL-Fab-HPC4 were pooled and concentrated by Millipore Amicon Ultra Centrifugal Filters (UFC 901096 10K NMWL, Billerica, Mass., USA). Purified protein was sterilized by filtration through a 0.2 mm filter unit (16541 Sartorius, Goettingen, Germany). The final protein concentration was determined by measuring 280 nm absorbance with a NANODROP UV spectrometer.

Purification of Human GFRAL Fragments Fusion Proteins

Five GFRAL fragments containing different domains were purified i.e. hGFRAL_C1_Fab_HPC4, hGFRAL_C2_Fab_HPC4, hGFRAL_C3_Fab_HPC4, hGFRAL_C1C2_Fab_HPC4 and hGFRAL_C2C3_Fab_HPC4.

For each GFRAL fragment fusion protein, culture supernatant containing secreted protein was applied to an anti-HPC4 sepharose 4FF affinity column (anti-HPC4 antibody coupled to the CNBr activated sepharose 4FF resin, 5 mL), equilibrated in 20 mM Tris-HCl, 100 mM NaCl, 1 mM CaCl2, pH 7.4. The bound protein was eluted with 20 mM Tris-HCl, 100 mM NaCl, pH 7.4, 1 mM EGTA. Fractions were pooled and concentrated to a final volume of approx. 3.0 mL using Millipore Amicon Ultra Centrifugal Filters (UFC 901096 10K NMWL, Billerica, Mass., USA) and further purified by size exclusion chromatography on a HiLoad 16/60 Superdex 200 μg column (28-9893-35 GE Healthcare, Uppsala, Sweden) in phosphate buffered saline (PBS). The fractions containing monomeric GFRAL fragment fusion protein were pooled and concentrated by Millipore Amicon Ultra Centrifugal Filters (UFC 901096 10K NMWL, Billerica, Mass., USA).

For hGFRAL_C3_Fab_HPC4, the protein was further polished by cation exchange chromatography. The fractions from size exclusion chromatography were pooled and diluted 10 fold into 20 mM NaAc pH5.0 and then applied to a Mono S 5/52 GL column (17-5168-01 GE Healthcare, Uppsala, Sweden).The bound protein was eluted with a 100-500 mM linear gradient of NaCl in 30 column volumes followed by a 600 mM NaCl step elution in 5 column volumes in 20 mM NaAc pH5.0. The charge variants of hGFRAL_C3_Fab_HPC4 were pooled separately and buffer-exchanged to PBS by Millipore Amicon Ultra Centrifugal Filters (UFC 901096 10K NMWL, Billerica, Mass., USA).

Purified protein was sterilized by filtration through a 0.2 mm filter unit (16541 Sartorius, Goettingen, Germany). The final protein concentration was determined by measuring 280 nm absorbance with a NANODROP UV spectrometer. Purified protein was analyzed by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and submitted to QC tests including endotoxin test and SEC-MALS. Protein identity was confirmed by LC-MS.

Example 5 Purification of Human RET Extracellular Region Fusion Proteins

Purification of hRET(ECR)-Fab-HPC4

The culture supernatant containing secreted hRET(ECR)-Fab-HPC4 was applied to an anti-HPC4 sepharose 4FF affinity column (anti-HPC4 antibody coupled to the CNBr activated sepharose 4FF resin, 5 mL), equilibrated in 20 mM Tris-HCl, 100 mM NaCl, 1 mM CaCl2, pH 7.4. The bound hRET(ECR)-Fab-HPC4 protein was eluted with 20 mM Tris-HCl, 100 mM NaCl, pH 7.4, 1 mM EGTA. Fractions were pooled and concentrated to a final volume of approx. 3.0 mL using Millipore Amicon Ultra Centrifugal Filters (UFC 901096 10K NMWL, Billerica, Mass., USA) and further purified by size exclusion chromatography on a Hi-Load 16/60 Superdex 200 μg column (28-9893-35 GE Healthcare, Uppsala, Sweden) in phosphate buffered saline (PBS). The fractions containing monomeric hRET(ECR)-Fab-HPC4 were pooled and concentrated by Millipore Amicon Ultra Centrifugal Filters (UFC 901096 10K NMWL, Billerica, Mass., USA). Purified protein was sterilized by filtration through a 0.2 mm filter unit (16541 Sartorius, Goettingen, Germany). The final protein concentration was determined by measuring 280 nm absorbance with a NANODROP UV spectrometer. Purified protein was analyzed by sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and submitted to QC tests including endotoxin test and SEC-MALS. Protein identity was confirmed by LC-MS.

Example 6 Throughput Plasmid Preparation for Cell Surface Display of Target Proteins

In order to get high-quality plasmids for cell surface display of target proteins on HEK293-6E cells by transient transfection, a high throughput method was designed and optimized to ensure that the extracted plasmids are free from contamination, and the endotoxin level in extracted plasmids is low enough for transfection of mammalian cells. The Nucleo-Bond® 96 Xtra EF kit (MACHEREY-NAGEL, Cat #: 740430.4) was used in plasmid extraction process with optimized protocol as described as follows.

At first, the stock plasmids were transformed into an E. coli strain, DH5α. Single colonies of each transformation were picked to inoculate a 1.0 mL start culture (LB medium) in 2.0 mL deep-well plates (Costar, Cat #: 3960). 100 ng/mL carbenicillin was used in the culture medium to give strong antibiotic selection pressure for retaining plasmids inside E. coli cells. The culture plates were incubated in a shaking incubator at 37° C., 800 rpm overnight. In order to have robust growth of the culture and enough culture volume for plasmid extraction, a two-step culture strategy was developed. The start culture after overnight growth was subcultured into 4 copies of 1.5 mL culture (TB medium, 100 ng/mL carbenicillin) in 2.0 mL deep-well plates which were incubated again in a shaking incubator at 37° C., 800 rpm overnight. So for each plasmid, 4×1.5=6 mL culture was prepared for extraction. Cells in each 1.5 mL culture plate were lysed independently, and 4 copies of cleared lysate were loaded to a single well of a plasmid DNA binding plate. The plasmid DNA binding plate was washed to remove impurities and endotoxin, and subsequently plasmids were eluted to a collection plate (Eppendorf, Cat #: 0030 502.140) using an endotoxin-free buffer. All liquid handling steps were automated using a liquid handling robot. The entire extraction process was optimized and tested so that there was no cross-well contamination, and the extracted plasmids were free of microbial contamination.

Example 7 Transient Transfection of HEK293-6E Cells to Display the Target Proteins on the Cell Surface Cells: HEK293-6E Reagents:

Gibco® FreeStyle™ 293 Expression Medium, Invitrogen, Cat. No.: 12338-026;

Gibco® 10% Pluronic F68, Invitrogen, Cat. No.: 24040-032;

Gibco® Geneticin, Invitrogen, Cat. No.: 11811-023;

Gibco® Opti-MEM® I+GlutaMax™-I, Invitrogen, Cat. No.: 51985-034;

293Fectin™ Reagent, Invitrogen, Cat. No.: 12347-019;

Equipment:

Infors HT Multitron

Tissue culture plate, 24 well, flat bottom with low evaporation lid (Falcon®, Cat. No.: 353047)

Plasmids:

ECR-LC (ECR-ATNP mIgG1 Fab LC) for TM1 like GFRAL

HC-TM (ATNP mIgG1 Fab HC-PDGFR TM) for TM1

LC-ECR (ATNP mIgG1 Fab LC-ECR) for TM2

TM-HC (OX40L TM-ATNP mIgG1-HC Fab) for TM2

Transient Expression:

This process is to transfect the HEK293-6E cells with corresponding plasmid DNA to display the ECR (extracellular region) of target protein on the surface of the cells as a fusion protein with the Fab of mouse anti-trinitrophenyl (ATNP) mAb. To achieve this, the plasmid encoding a protein fusion between the ECR and light chain of the Fab (ECR-LC or LC-ECR) was co-transfected into HEK293-6E cells with a plasmid encoding a protein fusion between the heavy chain of the Fab and a transmembrane domain (HC-TM or TM-HC). When expressed, the ECR and Fab light chain fusion protein formed a complex with the Fab heavy chain, which is anchored on the cell surface through the transmembrane domain.

The Fab light chain was located either at the N-terminus or at the C-terminus of the ECR of the target protein, depending on the transmembrane type of the target protein. If it is type 1 transmembrane protein (TM1), e.g. GFRAL, the Fab light chain is located at the C-terminus (ECR-LC) and the plasmid for co-transfection carries a fusion gene of Fab heavy chain and PDGFR transmembrane domain, namely HC-TM (ATNP mIgG1-Fab-PDGFR TM). If it is type 2 transmembrane protein (TM2), the Fab light chain is located at the N-terminus (LC-ECR) and the plasmid for co-transfection carries a fusion gene of OX40L transmembrane domain and Fab heavy chain, namely TM-HC (OX40L TM-ATNP mIgG1-HC Fab). The expression of ATNP mIgG1-Fab-PDGFR TM and OX40L TM-ATNP mIgG1-HC Fab was droved by CMV promoter in the vector. Type 1 transmembrane proteins are single-pass transmembrane proteins which have their N-terminus exposed to the extracellular space, while type 2 transmembrane proteins have their C-terminus exposed to the extracellular space.

Suspension HEK293-6E cells were grown with 5% CO2 and shaking at 37° C. in Gibco® FreeStyle™ 293 Expression Medium supplemented with 0.1% Fluronic F68 and 25 μg/ml of Geneticin. The cells were passaged between 0.3 and 2.5×106 cells/ml.

On the day of transfection, 1 ml of the cells with the density of 1.1×106 cells/ml and the viability of higher than 90% were prepared with fresh, pre-warmed cell culture medium (Gibco® FreeStyle™ 293 Expression Medium supplemented with 0.1% Fluronic F68 and 25 μg/ml of Geneticin) and allocated to 24we11 culture plate.

1 μg of plasmid DNA, including 0.8μg ECR-LC or LC-ECR DNA and 0.2μg HC-TM or TM-LC DNA, was diluted in Opti-MEM® I with GlutaMax™-I to a total volume of 33.3 ul. Meanwhile, 1 μl of 293Fectin™ Reagent was diluted in Opti-MEM® I with GlutaMax™-I to a total volume of 33.3 ul. The 293Fectin mixture was incubated for 5 minutes at room temperature. After the 5-minute incubation, the diluted 293Fectin™ Reagent was added to the diluted DNA, and further incubated for 20 minutes at room temperature to allow the 293fectin™-DNA complexes to form.

After the 293fectin™ and DNA incubation is complete, the 293fectin™-DNA mixture was added to the 1 ml HEK293-6E cells. The cells were incubated at 37° C. on an orbital shaker rotating at 250 rpm (orbit 25 mm) with 5% CO2. The cells were harvested at 40 hours after transfection and analyzed by FACS. The results are presented in both tables and figures, including curves and FACS dot-plot drawings.

Example 8 mAb26 Preparation and the In Vivo Activities Thereof

mAb26 is a monoclonal blocking antibody of wtMIC-1. The preparation and characterization of mAb26 can be found in Fairlie W D et al., Epitope mapping of the transforming growth factor-beta superfamily protein, macrophage inhibitory cytokine-1 (MIC-1): identification of at least five distinct epitope specificities, Biochemistry. 2001 Jan. 9; 40(1):65-73.

We confirmed that mAb26 can inhibit wtMIC-1's in vivo activity by reversing its food intake suppressing.

In the in vivo study, rats were pre-treated IP with 7 mg mAb26/animal and an isotype antibody as control (Ctrl. Ab). Without treatment of mAb26, wtMIC-1 reduced 24 hr food intake by about 40% in animals. In contrast, this food intake suppressing activity was almost abolished by mAb26. This study suggests that mAb26 blocks MIC-1's binding to MIC-1's functional receptor. Please see FIG. 1.

Example 9 Anti-MIC-1 Antibody Blocks Binding of MIC-1 to Soluble Human and Mouse GFRAL(ECR)-Fab-HPC4

A Biacore® 4000 (GE Healthcare, Piscataway, N.J., USA) instrument was used for SPR-based blocking assay to analyze the binding specificity between wild type human MIC-1 (wtMIC-1) and GFRAL. The assays were performed at 25° C. at flow rates of 30 μL/minute in 1× HBS-P running buffer (BR-1006-71, GE Healthcare, Piscataway, N.J., USA). wtMIC-1 was mixed with various concentration of MIC-1 blocking antibody mAb26 to form complex of wtMIC-1 and mAb26. The mixture was injected as analyte. hGFRAL(ECR) was immobilized on the surface of the chip as ligand. wtMIC-1 binding to immobilized hGFRAL(ECR) was detected. With the decreased binding level (response unit, RU), mAb26 dose-dependently blocked wtMIC-1's binding to immobilized hGFRAL(ECR) whereas isotype control (mIgG1) did not (FIG. 2, Table 1). This further suggested the binding of MIC-1 to hGFRAL was specific.

TABLE 1 Binding of wtMIC-1 to hGFRAL(ECR) was blocked by mAb26 in SPR Response (RU) Ab Conc. (nM) wtMIC-1 + mAb26 wtMIC-1 + mIgG1 (ctrl.) 0 57.1 58.3 0.125 52.1 58.4 0.25 47 59.4 0.5 35.4 58.4 1 13.8 59.3 2 1.8 60 4 1.6 59.1 8 1.9 59.4

Similarly, the binding of MIC-1 to mouse GFRAL was characterized with Biacore® 4000. wtMIC-1 was mixed with various concentration of mAb26 to form complex and injected as analyte; while mGFRAL(ECR) was immobilized as ligand. wtMIC-1 binding to immobilized mGFRAL(ECR) was detected. With the decreased binding level (response unit, RU), mAb26 dose-dependently blocked wtMIC-1 binding to immobilized mGFRAL(ECR) whereas isotype control did not. This further suggested the binding between MIC-1 and GFRAL is specific (FIG. 3, Table 2).

TABLE 2 Binding of wtMIC-1 to mGFRAL(ECR) was blocked by mAb26 in SPR Response (RU) Ab Conc. (nM) wtMIC-1 + mAb26 wtMIC-1 + mIgG1 (ctrl.) 0 56.2 55.6 0.125 49.6 55.3 0.25 44.2 54.6 0.5 31.7 55 1 8.9 54.1 2 0.8 54.5 4 0.9 55.4 8 0.2 55.5

Example 10 Ret Binds to Complex of MIC-1 and GFRAL

hRet and complex of hGFRAL and MIC-1 interactions were assessed by SPR-based binding assay. A Biacore® 4000 (GE Healthcare, Piscataway, N.J., USA) instrument was used. The assays were performed at 25° C. at flow rates of 30 μL/minute in 1× HBS-P running buffer (BR-1006-71, GE Healthcare, Piscataway, N.J., USA). hRet(ECR)-Fc was immobilized through Fc as ligand on the chip. VISTA-Fc was immobilized as negative control ligand. The complex of MIC-1 and hGFRAL was prepared by pre-incubating wtMIC-1 with hGFRAL(ECR)-His. wtMIC-1, hGFRAL(ECR)-His, complex of wtMIC-1 and Ctrl-His, and complex of wtMIC-1 and hGFRAL(ECR)-His at various concentration were run as flow-through analyte.

Results showed that hRet only bound to complex of wtMIC-1 and hGFRAL(ECR)-His, but not to any other analytes (FIG. 4 and Table 3). VISTA did not bind to any of the analytes. Thus, it can be concluded that that hRet binds to hGFRAL in the presence of MIC-1; and Ret, MIC-1 and GFRAL forms a ternary complex.

TABLE 3 Ret bound to complex of MIC-1 and GFRAL Response (RU) Analyte Analyte Conc (nM) hRet-Fc Ctrl-Fc hGFRAL(ECR)-His 0 0 0 12 −0.03970337 −2.343414 37 −0.1051636 −2.039063 111 0.208313 −2.940094 333 −0.4967651 −3.480164 1000 −1.109711 −3.848938 wtMIC-1 0 0 0 12 0.1835941 −0.244781 37 0.4674381 −0.017883 111 0.4840397 0.361664 333 0.8801884 0.225617 1000 0.5267033 −0.514648 wtMIC-1 + Ctrl-His 0 0 0 12 −0.179032 −0.1569062 37 −0.10286 −0.5586091 111 0.291336 −0.3733826 333 0.544601 −1.0348359 1000 0.324859 0.0764923 wtMIC-1 + 0 0 0 hGFRAL(ECR)-His 12 0.2256165 −1.3935545 37 1.9661557 −1.4817815 111 5.4817807 −1.018219 333 8.3245547 −1.4296875 1000 10.7024807 −1.4036565

Example 11 Survey of Binding Interactions Between GDNF Family (MIC-1, GDNF, NRTN, ARTN and PSPN) and GFRAL

For SPR-based binding assay, wtMIC-1 as well as hGDNF, hNRTN, hARTN and hPSPN, at various concentrations were run as flow-through analytes. hGFRAL(ECR)-Fab and mGFRAL(ECR)-Fab were immobilized as ligand. Results show that only wtMIC-1 bound to hGFRAL(ECR)-Fab and mGFRAL(ECR)-Fab. FCRL2-Fab-HPC4 (Ctrl.-Fab-HPC4) was used as control. Please see FIG. 5 and Table 4. Results showed that hGFRAL or mGFRAL only bound to wtMIC-1, but not to the other four ligands of GDNF family.

TABLE 4 hGDNF, hNRTN, hARTN or hPSPN did not bind to wtMIC-1 in SPR hGFRAL(ECR)- Ctrl.-Fab- mGFRA(ECR)L- Fab-HPC4 HPC4 Fab-HPC4 wtMIC-1 x hGDNF x x x hNRTN x x x hARTN x x x hPSPN x x x “✓”: binding was detected; “x”: No binding was detected

Example 12 Characterization of GFRAL Domain(s) for MIC-1 Interaction

The ECR of hGFRAL has three cysteine-rich domains: C1, C2 and C3, according to sequence comparison to GDNF family receptor α (GFRα) and secondary structure analysis. To identify which domain is responsible of binding to MIC-1, soluble GFRAL fragments (hGFRAL(ECR)-Fab-HPC4, hGFRAL_C1_Fab_HPC4, hGFRAL_C2_Fab_HPC4, hGFRAL_C3_Fab_HPC4, hGFRAL_C1C2_Fab_HPC4, and hGFRAL_C2C3_Fab_HPC4) were prepared according to previous examples.

MIC-1 interactions with soluble GFRAL fragments were assessed by SPR-based binding assay. A Biacore® 4000 (GE Healthcare, Piscataway, N.J., USA) instrument was used for SPR-based binding assay. The assays were performed at 25° C. at flow rates of 30 μL/minute in 1× HBS-P running buffer (BR-1006-71, GE Healthcare, Piscataway, N.J., USA). Purified GFRAL fragments at various concentrations were run as flow-through analyte and wtMIC-1 was immobilized as ligand. Please see FIG. 6 and Table 5.

TABLE 5 Relative binding of wtMIC-1 to GFRAL fragments Ligand % of Binding hGFRAL(ECR)-Fab-HPC4 100 hGFRAL_C1_Fab_HPC4 N.A. hGFRAL_C2_Fab_HPC4 21 hGFRAL_C3_Fab_HPC4 N.A. hGFRAL_C1C2_Fab_HPC4 87 hGFRAL_C2C3_Fab_HPC4 8 *Binding between wtMIC-1 and hGFRAL(ECR) was detected as the strongest, thus was set up as 100% “N.A.”: not available

According to the results shown above, hGFRAL_C1C2 showed strong binding to wtMIC-1. In addition, hGFRAL_C2 and hGFRAL_C2C3 at 1000 nM showed weak but apparent binding to immobilized wtMIC-1 in SPR, which indicates that C2 involves in direct binding with MIC-1 and should be the crucial fragment.

Example 13 Characterization of GFRAL Domain(s) for Ret to Bind with MIC-1/GFRAL Complex

To identify which domain of GFRAL (C1, C2 and C3) is responsible of MIC-1/GFRAL/Ret ternary complex formation, flow cytometry-based binding assay was performed. Binding of Ret-Fc protein against HEK cells expressing hGFRAL(ECR) and its fragments (C1, C2 and C3) was tested with presence or absence of wtMIC-1 through FACS.

HEK293-6E cells were transiently transfected with plasmid expressing hGFRAL(ECR) and its fragments (hGFRAL_C1, hGFRAL_C2, hGFRAL_C3, hGFRAL_C1C2, hGFRAL_C2C3) were stained with Ret-Fc protein with the presence or absence of wtMIC-1. GFRAL fragments expression on cell surface was verified by positive staining of anti-mouse-Kappa antibody via flow cytometry analysis. Binding of hRet(ECR)-Fc was detected using APC-conjugated goat Anti-Human IgG (13392/D3-110, Cayman Chemical). Results showed that when there was no wtMIC-1, RET did not bind to hGFRAL(ECR) or its fragments (hGFRAL_C1, hGFRAL_C2, hGFRAL_C3, hGFRAL_C1C2, hGFRAL_C2C3). When wtMIC-1 was present, RET only bound to hGFRAL(ECR) but no other fragments (hGFRAL_C1, hGFRAL_C2, hGFRAL_C3, hGFRAL_C1C2, hGFRAL_C2C3). Considering data in the previous example, i.e., both hGFRAL(ECR) and hGFRAL_C1C2 showed detectable binding to wtMIC-1, and RET bound to the complex of wtMIC-1 and GFRAL, the results of this example indicate that C3 domain is necessary for Ret's binding to the complex of MIC-1 and GFRAL, although C3 is dispensable for MIC-1's binding to GFRAL. Please see FIG. 7, Table 6; and FIG. 8 (showing the relative binding of Ret-Fc* in FIG. 7) and Table 7.

TABLE 6 Meaning of the four quadrants in the FACS dot-plot drawings of FIG. 7 Bio-MIC-1 binding hRet-Fc binding Q1 (upper left) + Q2 (upper right) + + Q3 (lower right) + Q4 (lower left)

TABLE 7 Relative binding of Ret-Fc* of FIG. 8 hGFRAL_ hGFRAL_ hGFRAL_ hGFRAL_ hGFRAL_ hGFRAL C1 C2 C3 C1C2 C2C3 (ECR) w/o probe 10 0 4 12 2 8 wtMIC-1 4 0 3 15 12 5 hRet-Fc 5 0 6 8 22 6 wtMIC-1 + hRet-Fc 6 12 6 5 16 100 *Binding between hRet-Fc and hGFRAL(ECR) with presence of wtMIC-1 was detected as the strong-est, thus was set up as 100%

Overall, data in Examples 15 and 16 indicates that GFRAL-C1C2 is critical for MIC-1 binding and GFRAL-C3 is essential for Ret binding to from MIC-1/GFRAL/Ret ternary complex.

Example 14 Generation of Stably-Transformed Mammalian Cells Expressing Both Full Length Human GFRAL and Full Length Human RET Cell Line: PC-12, HEK293 Expression Constructs

pEL-Full Length-hGFRAL

Human (Homo sapiens) GDNF family receptor alpha like (hGFRAL) DNA sequence (SEQ ID: 1) is from CCDS4957.1. The DNA sequence encoding the full length GFRAL protein was optimized to remove common restriction sites (EcoRI, BamHI, HindIII, and ApaI) while retaining the GFRAL peptide sequence.

hGFRAL DNA was synthesized and cloned into expression vector pEL, which contains a CMV promoter to drive expression in the mammalian cells.

Rat GFRAL (amino acid sequence is shown in SEQ ID NO: 22), cyno GFRAL (amino acid sequence is shown in SEQ ID NO: 20) and mouse GFRAL (amino acid sequence is shown in SEQ ID NO: 4) expression constructs were generated in a similar way as human GFRAL expression construct as described above, by cloning rat, cyno and mouse GFRAL cDNA sequences into expression vector pEL.

pEL-Full length-hRET51

Human (Homo sapiens) proto-oncogene tyrosine-protein kinase receptor's (RET51) DNA sequence is from CCDS7200.1 and UniProt P07949-1 (hRET51, amino acid sequence is shown in SEQ ID NO: 5). The DNA sequence encoding the full length GFRAL protein was optimized to remove common restriction sites (EcoRI, BamHI, HindIII, and ApaI) while retaining the RET peptide sequence.

hRET51 DNA was synthesized and cloned into expression vector pEL, which contains a CMV promoter to drive expression in the mammalian cells.

hRET43 (amino acid sequence is shown in SEQ ID NO: 17), hRET9 (amino acid sequence is shown in SEQ ID NO: 18), rat RET(amino acid sequence is shown in SEQ ID NO: 23), cyno RET (amino acid sequence is shown in SEQ ID NO: 21) and mouse RET (amino acid sequence is shown in SEQ ID NO: 19) expression constructs were generated in a similar way as human RET51 expression construct as described above, by cloning rat, cyno and mouse RET cDNA sequences into expression vector pEL. Please note that Rat RET, cyno RET and mouse RET are homolog of hRET51.

Reagent

DMEM (Gibco 10569)

Geneticin(Gibco 10131035)

Hygromycin B (Roche 10843555001)

Lipofectamine 2000 (Invitrogen 11668027)

Process:

    • 1) Seeded 4E6 PC-12 or HEK293 cells in 10 cm dish for O/N.
    • 2) Transfected PC-12 or HEK293 cells with 24 ug pEL-FL-hGFRAL, or pEL-FL-hRET51/43/9, or both pEL-FL-hGFRAL and pEL-FL-hRET51/43/9 in 60 ul Lipofectamine 2000 and culture at 37° C. for O/N.
    • 3) Split the original plate to daughter plates at different ratios 1:10 (2), 1:100 (2), 1:500 (2) and 1:1000 (2),
    • 4) 6 h after, added 10 mL DMEM (containing 2 mg/mi G418) to each dishes, the geneticin concentration in dishes was 1 mg/ml.
    • 5) Cells were collected and mRNA was purified with QIAGEN RNeasy mini kit; cDNA was obtained by Reverse Transcription with Thermoscript RT-PCR system; qPCR with AB Power SYBR Green PCR Master Mix was carried out to test transfection efficiency and the cDNA were kept as positive control for further clones validation use.
    • 6) Cells were with 1 mg/ml geneticin in DMEM for 3 weeks
    • 7) Single clones were picked up to 24 well plate and cells were grew till full confluence
    • 8) Expression of full length-hGFRAL and full length hRET51/43/9 were confirmed by western blot (Sigma, cat #HPA047372), and by qPCR (5′-gaatctaactacacgttcccatca-3′ and 5′-cagaccacatcccctacaca-3′) using the same procedure as above.
    • 9) Positive clones were further validated by functional assay

Example 15 Studying of MIC-1 signaling transduction Reagents

DMEM (Gibco 10569)

Hygromycin B (Roche 10843555001)

Trypsin: T4424, Sigma

D-PBS: D8537, Sigma

P/S: 15140-122, Gibco

RIPA buffer (Thermo, cat #89900)

Anti-Ret (phospho Y1062) antibody (Abcam, cat #ab51103)

Phospho-Ret (Tyr905) Antibody (Cell Signaling, cat #3221S)

Phospho-Met (Tyr1234/1235) (D26) XP® Rab (Cell Signaling, cat #3077S)

Phospho-Met (Tyr1003) (13D11) Rabbit mAb (Cell Signaling, cat #3135S)

Phospho-p38 MAPK (Thr180/Tyr182) (D3F9) XP® Rabbit mAb (Cell Signaling, cat #4511)

Phospho-SAPK/JNK (Thr183/Tyr185) (81E11) Rabbit mAb (Cell Signaling, cat #4668)

Phospho-EGF Receptor (Tyr1068) (D7A5) XP® Rabbit mAb (Cell Signaling, cat #3777)

AlphaScreen SureFire ERK1/2 (p-T202/Y204) Assay Kit (PerkinElmer, Catalog #TGRES10K)

AlphaScreen SureFire Akt1/2/3 (p-5473) Assay Kit (Perkin Elmer, Catalog #TGRA4S10K)

AlphaScreen SureFire SMAD1(p-5463/465) Assay Kit (PerkinElmer, Catalog #TGRSM1S10K)

AlphaScreen SureFire SMAD3(p-5423/425) Assay Kit (PerkinElmer, Catalog #TGRSM3S10K)

PC12, HEK293 cells were transfected with full length human GFRAL, or full length human RET51/43/9, or co-transfected with full length human GFRAL and full length human RET51/43/9 using Lipfectamine LTX (Life technologies, 15338100) according to previous example. Twenty-four hours after transfection, cells were seeded on 96 well plates at 1E6/ml. Twenty-four hours later, cells were starved in serum free culture medium for 4 hrs and treated with wild type human wtMIC-1 as indicated for 15 mins. Cells were lysed by Surefire lysis buffer and ERK1/2 phosphorylation (pERK1/2) was measured by Surefire a-screen. Results showed that MIC-1 only induced ERK1/2 phosphorylation in cells co-transfected with full length human GFRAL and full length human RET51, but not in cells transfected with GFRAL only, or in cells transfected with RET51 only. Please see FIG. 9 and Table 8; FIG. 10 and Table 9; and FIG. 11 and Table 10. Results also showed that MIC-1 only induced pERK in cells co-transfected with hRET51 and hGFRAL, not in cells co-transfected with hRET43 and hGFRAL or hRET9 and hGFRAL. Please see FIG. 12 and Table 11.

TABLE 8 wtMIC-1 induced ERK1/2 phosphorylation in PC12 cells expressing hGFRAL(FL) and hRET51(FL) wtMIC-1 log(nM) pERK1/2 (Evision Unit) 0.69897 800753.8 0.30103 796830.5 −0.09691 739949.0 −0.49485 709877.5 −0.89279 611715.2 −1.29073 532987.2 −1.68867 496081.5 −2.50000 564097.8

Results in Table 8 and FIG. 9 showed that wtMIC-1 dose dependently activated ERK1/2 phosphorylation in hGFRAL and hRET51 co-transfected PC12 cells with an EC50 around 0.2 nM.

TABLE 9 wtMIC-1 induced ERK1/2 phosphorylation in HEK293 cells co-transfected with hGFRAL(FL) and hRET51(FL) wtMIC-1 (nM) pERK1/2 (Evision Unit) 100.000000 3565 25.000000 3569 6.250000 4215 1.562500 4522 0.390625 4361 0.09765625 3529 0.02441406 2632 0.006103516 2321

Results in Table 9 and FIG. 10 showed that wtMIC-1 dose dependently activated ERK1/2 phosphorylation in hGFRAL and hRET51 co-transfected HEK293 cells.

TABLE 10 wtMIC-1 did not induce ERK1/2 phosphorylation in PC-12 cells expressing only hGFRAL(FL) or only hRET51(FL) pERK1/2 (Evision Unit) wtMIC-1 no- hGFRAL hRET51 hGFRAL/hRET51 (nM) ransfection (wt) transfected transfected co-transfected 100.00000 96752.3 60239.2 66502.2 393718.7 33.33330 93058.5 51846.0 59253.7 371121.5 11.11110 105186.3 53627.0 65703.5 385059.0 3.70370 96438.67 56083.2 63068.2 375673.0 1.23460 100130.8 61954.2 65140.5 361845.5 0.41150 102684.8 61078.3 64341.7 303567.2 0.13720 108993.7 57031.0 65766.3 213397.5 0.04570 102958.8 57841.7 67729.7 163937.7 0.01520 106936.3 52780.5 64593.2 151551.3 0.00200 109256.7 49181.2 66674.5 146359.7

Results in Table 10 and FIG. 11 showed that wtMIC-1 did not induce ERK1/2 phosphorylation in PC12 cells transfected with only hGFRAL or hRET51, but only cells co-transfected with both hGFRAL and hRET51. Thus, both GFRAL and RET are need for MIC-1 to have activity.

TABLE 11 wtMIC-1 did not induce pERK in HEK293 cells co-transfected with RET43/9 and hGFRAL, but pERK only in HEK293 cells co-transfected with RET51 and hGFRAL pERK1/2 (Evision Unit) wtMIC-1 (nM) RET 9 RET43 RET51 66.666670 1.323266 1.158630 1.737150 22.222220 1.255731 1.105068 1.727052 7.407407 1.302666 1.136891 1.701729 2.469136 1.244610 1.072073 1.686027 0.823045 1.170909 1.090099 1.395468 0.274348 1.069708 1.051677 1.132260 0.091449 0.988070 1.009341 0.969390 0.030483 1.066978 1.034423 1.023812 0.001000 1.000000 1.000000 1.000000

Example 16 Cross-Species Reaction of MIC-1

BHK cells were co-transfected with full length rat GFRAL and rat RET (homolog of hRET51), or cyno GFRAL and cyno RET (homolog of hRET51) using Lipfectamine LTX (Life technologies, 15338100) according to previous example. Twenty-four hours after transfection, cells were seeded on 96 well plates at 1E6/ml. Twenty-four hours later, cells were starved in serum free culture medium for 4 hrs and treated with wild type human wtMIC-1 as indicated for 15 mins. Cells were lysed by Surefire lysis buffer and ERK1/2 phosphorylation (pERK1/2) was measured by Surefire a-screen.

Results showed that MIC-1 also induced ERK1/2 phosphorylation in cells co-transfected with rat GFRAL and rat RET, or cells co-transfected with cyno GFRAL and cyno RET. This means that human MIC-1 has cross-species activity, which would significantly facilitate drug development, since the same MIC-1 compound could be applied to animals in pre-clinical trials and clinical trials. Please see FIG. 13 and Table 12.

TABLE 12 wtMIC-1 induced pERK in rat and cyno GFRAL/RET transfected BHK21 cells pERK1/2 (Evision Unit) wtMIC-1 (nM) rat GFRAL/RET Cyno GFRAL/RET 100.000000 40572.00 63944.25 25.000000 37384.75 65745.25 6.250000 40738.75 63194.50 1.562500 47997.75 64888.25 0.390625 48433.75 37388.75 0.09765625 23426.75 24647.75 0.02441406 17716.50 21785.25 0.006103516 17309.50 20982.50 0.001525879 172810 19753.50 0.000100 18255.50 19986.25

Results in Table 12 showed that cross-species activity of human wtMIC-1 with rat GFRAL/RET and cyno GFRAL/RET.

Based on the results above, we believe GFRAL is the cell surface receptor that mediates the in vivo activities of MIC-1. In particular, GFRAL binds to MIC-1, then RET binds to the complex of GFRAL and MIC-1 to form ternary complex. Such ternary complex phosphorylates RET protein tyrosine kinase to induce in vivo activities of MIC-1 through signal pathways comprising at least comprising ERK/MAPK pathway by phosphorylation of ERK1/2 pathway.

Example 17 Further Studying of MIC-1 Signaling Transduction

In order to profile MIC-1 induced signaling systematically, MIC-1 function on GFRAL/RET co-transfected BHK21 cells was examined by Cignal 45-pathway following the manufacturer's protocol.

Reagent:

Cignal 45-pathway Reporter Array (Qiagen: CCA-901L)

Dual-Glo® Luciferase Assay System (Promega: E2940)

Briefly, BHK21 cells were seeded on Cignal 45-pathway plates and transfected with hGFRAL and hRET plasmids as described above. The transfected were treated with 10 nM wild type hMIC-1 for 4 hours and Luciferase signal was quantified.

Cignal 45-pathways results showed that Hedgehog, MEF2 and MAPK/ERK signaling pathways were regulated upon MIC-1 treatment, as evidenced by significant upregulation of GLI, MEF2 and SRE derived luciferase reporter activities respectively (according to 1 way ANOVA, p<0.05 for MIC-1 vs. inactive MIC-1 analogue). Please see FIG. 14.

However, we did not observe the MIC-1 induced FOXO activity (corresponding to the pathway comprising AKT1/2/3 phosphorylation) in this experiment, which we believe was due to that MIC-1 response window or assay sensitivity of this specific pathway is not sufficient to be observed by this assay.

Furthermore, regulation of GLI and MEF2 signaling was confirmed by quantification of GLI and MEF2 mRNA expression in wtMIC-1 treated BHK21 cells by qPCR.

Example 18 Establishment of BHK21-hGFRAL-IRES-hRET-SRE-Luc Stable Cells

The purpose of this example was to establish a cell based in vitro assay for testing MIC-1 activity. Mammalian cells were transfected and stably expressed full length hGFRAL and full length hRET51; and the activity of MIC-1 can be examined by detecting luciferase activity.

Cell line: BHK21 Cells (ATCC: tk-ts13)

Plasmid

pEL-hGFRAL-IRES-hRET

Plasmids expressing full length hGFRAL and full length hRET51 were constructed by inserting synthesized DNA nucleotides encoding full length hGFRAL and full length hRET51 into mammalian expression vector pEL. IRES (internal ribosome entry site) is a commonly used linker between two DNA sequences, so that the two DNA sequences can be simultaneously translated into mRNA. pEL vector backbone was provided by Taihegene CRO company.

pGL4.33-SRE-Luc

Plasmids expressing luciferase were constructed by inserting synthesized DNA sequence of luciferase into mammalian expression vector pGL4.33. SRE is the abbreviation of “serum response element”. When MIC-1 binds to GFRAL and RET, ERK is phosphorylated as pERK. pERK can increase the expression of serum response factor, and serum response factor binds to SRE to up regulate the expression of luciferase, so that MIC-1 activity can be tested and indicated as the activity of luciferase. pGL4.33 vector backbone was provided by Taihegene CRO company.

Reagent

DMEM (Gibco 10569)

Geneticin(Gibco 10131035)

Hygromycin B (Roche 10843555001)

Lipofectamine 2000 (Invitrogen 11668027)

Process: Generation of BHK21-hGFRAL-IRES-hRET Stable Cells

Two millions of BHK21 cells were seeded in a 10 cm petri dish and cultured for overnight in culture medium (DMEM+10% FBS+1% PS). Cells were transfected with pEL-hGFRAL-IRES-hRET plasmids. Transfected cells were split into new 10 cm dishes at different densities and grew in selection medium (DMEM+10% FBS+1% PS+1 mg/ml G418) for more than 2 weeks to get single clones. The single clones were transferred to 6 well plates and cultured to 100% confluence. mRNA expression of hGFRAL and hRET was measured by qPCR. Successfully transfected clones were picked up and tested for MIC-1 binding (FIG. 15 and Table 13). MIC-1 induced both phosphorylation of ERK and AKT in BHK21-hGFRAL-IRES-hRET stable cells. Please see FIG. 16, FIG. 17 and Table 14, and FIG. 18 and Table 15. The results show that the ternary complex of MIC-1, GFRAL and RET phosphorylates RET protein tyrosine kinase to induce in vivo activities of MIC-1 through signal pathways comprising not only ERK/MAPK pathway by phosphorylation of ERK1/2 pathway, but also PI3K/Akt pathway by phosphorylation of AKT1/2/3.

One clone that had the best RET/ERK/AKT phosphorylation level was selected for further transfection of pGL4.33-SRE-Luc plasmids.

TABLE 13 wtMIC-1 binds to BHK21-hGFRAL-IRES-hRET stable cells (EC50 = 1.3 nM) wtMIC-1 binding wtMIC-1 (nM) (MIF: median fluorescence intensity) 100 926 33.33333333 924 11.11111111 882 3.703703704 816 1.234567901 625 0.411522634 347 0.137174211 333 0.045724737 400 0.015241579 316 0.001 300

TABLE 14 wtMIC-1 induced ERK phosphorylation in BHK21-hGFRAL-IRES-hRET stable cells ERK1/2 phosphorylation wtMIC-1 (nM) (alpha signal (counts)) 100 436226 25 429956 6.25 442116 1.5625 308222 0.390625 72323 0.097656 41955 0.024414 42171 0.006104 35645 0.001526 40198 0.0001 44930

TABLE 15 wtMIC-1 induced AKT phosphorylation in BHK21-hGFRAL-IRES-hRET stable cells AKT1/2/3 phosphorylation wtMIC-1 (nM) (alpha signal (counts)) 100 141354 25 129392 6.25 145118 1.5625 107735 0.390625 58760 0.097656 40976 0.024414 35777 0.006104 33394 0.001526 43367 0.0001 41320

Generation of BHK21-hGFRAL-IRES-hRET-SRE-Luc Stable Cells

The selected clone of BHK21-hGFRAL-IRES-hRET cells was grown to two million. Then the two million BHK21-hGFRAL-IRES-hRET cells were seeded in a 10 cm petri dish and cultured in culture medium 2 (DMEM+10% FBS+1% PS+1 mg/ml G418). Cells were transfected with pGL4.33-SRE-Luc plasmids (seq.2) following the standard protocol. Transfected cells were split into new 10 cm dishes at different densities and grew in selection medium (DMEM+10% FBS+1% PS+1 mg/ml G418+hygromycin 400 μg/ml) for more than 2 weeks to get single clones. The single clones were transferred to 6 well plates and cultured to 100% confluence. Clones were tested for induction of luciferase activity upon wtMIC-1 treatment. Clones that showed high luciferase activity induced by wtMIC-1 were cultured and cryopreserved.

MIC-1 Induced ERK Phosphorylation in BHK21-hGFRAL-IRES-hRET-SRE-Luc Stable Cells

BHK21-hGFRAL-IRES-hRET-SRE-Luc stable cells were seeded at 2E4 cells per well in 96-well plates and cultured for overnight. Medium was removed and replenished with 100 μl serum free DMEM for 4 hours. Different concentrations of wtMIC-1 were added to the medium and incubated for 15 minutes. ERK1/2 was quantified by ERK1 (p-Thr 202) and ERK2 (p-Thr204) alphascreen Surefire assay kits following manufacturer instructions. Dose response curves were generated by nonlinear regression curve fit (4-parameters) with GraphPad Prism. Results were shown in FIG. 19 and Table 16.

TABLE 16 wtMIC-1 induced ERK1/2 phosphorylation in BHK21-hGFRAL-IRES-hRET-SRE-Luc stable cells Control (inactive MIC-1 ERK1/2 phosphorylation analogue) wtMIC-1 (nM) (alpha signal (counts)) (alpha signal (counts)) 100 134920.5 5502.5 28.57143 127624.5 5331.0 8.163265 130249.0 5783.5 2.332362 126250.5 5111.5 0.666389 90359.0 4970.0 0.190397 32908.5 6292.5 0.054399 7851.0 5058.5 0.015543 5010.0 5388.5 0.004441 4751.5 N/A 0.001 4736.0 N/A

MIC-1 Activated Luciferase in BHK21-hGFRAL-IRES-hRET-SRE-Luc Stable Cells

BHK21-hGFRAL-IRES-hRET-SRE-Luc stable cells were seeded at 4E4 cells per well in 96-well plates and 6 hours after cell seeding, medium was removed and replenished with 100 μl serum free DMEM for overnight. Different concentrations of wtMIC-1 were added to the medium and incubated for 4 hours. Steady light plus (PE 6016751) was added to the plates and incubated for 30 min at room temperature. The signal was measured in Envision, ultra luminescence and dose response curves were generated by nonlinear regression curve fit (4-parameters) with GraphPad Prism. Results were shown in FIG. 20 and below Table 17.

TABLE 17 wtMIC-1 activated luciferase in BHK21-hGFRAL- IRES-hRET-SRE-Luc stable cells Control (inactive MIC-1 Luminescence activity analogue) wtMIC-1 (nM) (alpha signal (counts)) (alpha signal (counts)) 62.5 60700 28380 15.625 56800 23580 3.90625 55140 24780 0.976563 46560 22940 0.244141 24720 22000 0.061035 19220 21540 0.015259 18380 22960 0.001 18220 22860

While certain features of the invention have been illustrated and described herein, many modifications, substitutions, changes, and equivalents will now occur to those of ordinary skill in the art. It is, therefore, to be understood that the appended claims are intended to cover all such modifications and changes as fall within the true spirit of the invention.

Claims

1. A method for determining the activity of a MIC-1 compound, comprising:

(a) contacting an indicator cell with a test sample that comprises the MIC-1 compound; and
(b) detecting a biological response of the indicator cell contacted with the test sample;
wherein the indicator cell expresses on the cell surface a MIC-1 binding receptor comprising an amino acid sequence that is at least 80% identical to the human GDNF family receptor alpha like (GFRAL) amino acid sequence set forth in SEQ ID NO:1.

2. The method of claim 1, wherein the amino acid sequence of the MIC-1 binding receptor is SEQ ID NO:1.

3. The method of claim 1, wherein the indicator cell further expresses a cell surface receptor kinase, and wherein the biological response is induced when the MIC-1 compound, the MIC-1 compound binding receptor, and the cell surface receptor kinase form a ternary complex.

4. The method of claim 3, wherein the cell surface receptor kinase is RET receptor tyrosine kinase.

5. The method according to claim 1, wherein the MIC-1 binding receptor comprises the C1, C2, and C3 domains of the human GFRAL.

6. The method according to claim 1, wherein the biological response is an increase or decrease of phosphorylation of a protein kinase.

7. The method of claim 6, wherein the protein kinase is ERK1/2 or AKT1/2/3.

8. An in vitro cell line for determining the activity of a MIC-1 compound, wherein the cell line expresses a MIC-1 binding receptor and a cell surface receptor kinase, wherein the MIC-1 binding receptor comprises an amino acid sequence that is at least 80% identical to the human GDNF family receptor alpha like (GFRAL) amino acid sequence set forth in SEQ ID NO:1, wherein the cell line expresses a reporter peptide, and wherein a biological response is induced when the MIC-1 compound, the MIC-1 binding receptor, and the cell surface receptor kinase form a ternary complex.

9. The cell line of claim 8, wherein the cell surface receptor kinase is RET receptor tyrosine kinase.

10. (canceled)

11. The cell line of claim 8, wherein the reporter peptide is luciferase.

12. The cell line of claim 1, wherein the indicator cell is selected from the group consisting of BHK21 cell, PC-12 cell, HEK293 cell, or HEK293 cell stably expressing the Tet repressor.

13. An in vitro cell line for determining the activity of a MIC-1 compound, wherein the cell line expresses a human GDNF family receptor alpha like (GFRAL) comprising the amino acid sequence set forth in SEQ ID NO:1, a hRET comprising the amino acid sequence set forth in SEQ ID NO:5 and luciferase.

14. A method for detecting the binding affinity of a MIC-1 compound and human GDNF family receptor alpha like (GFRAL), comprising:

(a) contacting a test sample comprising the MIC-1 compound with a MIC-1 binding protein; and
(b) detecting binding of between the MIC-1 compound and the MIC-1 binding protein, wherein the MIC-1 binding protein comprises the C1 and C2 domains of GFRAL.
Patent History
Publication number: 20190025332
Type: Application
Filed: Jan 13, 2017
Publication Date: Jan 24, 2019
Patent Grant number: 11105818
Inventors: Jing Su (Beijing), Wei Yang (Boston, MA), Chih-Chuan Chang (Beijing), Li Yang (Beijing), Zhe Sun (Beijing), Haibin Chen (Ningbo), Zhenhua He (Shanghai), Zhe Wang (Beijing), Sebastian Beck Joergensen (Virum)
Application Number: 16/069,985
Classifications
International Classification: G01N 33/74 (20060101); C07K 14/71 (20060101); C12N 9/12 (20060101);